the endocrine system in sports and exercise library...the endocrine system in sports and exercise /...

646
THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE VOLUME XI OF THE ENCYCLOPAEDIA OF SPORTS MEDICINE AN IOC MEDICAL COMMISSION PUBLICATION IN COLLABORATION WITH THE INTERNATIONAL FEDERATION OF SPORTS MEDICINE EDITED BY WILLIAM J. KRAEMER AND ALAN D. ROGOL

Upload: others

Post on 24-Feb-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE

VOLUME XI OF THE ENCYCLOPAEDIA OF SPORTS MEDICINE

AN IOC MEDICAL COMMISSION PUBLICATION

IN COLLABORATION WITH

THE INTERNATIONAL FEDERATION OF SPORTS MEDICINE

EDITED BY

WILLIAM J. KRAEMER AND ALAN D. ROGOL

Page 2: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 3: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE

Page 4: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 5: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE

VOLUME XI OF THE ENCYCLOPAEDIA OF SPORTS MEDICINE

AN IOC MEDICAL COMMISSION PUBLICATION

IN COLLABORATION WITH

THE INTERNATIONAL FEDERATION OF SPORTS MEDICINE

EDITED BY

WILLIAM J. KRAEMER AND ALAN D. ROGOL

Page 6: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

© 2005 International Olympic CommitteePublished by Blackwell Publishing LtdBlackwell Publishing, Inc., 350 Main Street, Malden, Massachusetts 02148-5020, USABlackwell Publishing Ltd, 9600 Garsington Road, Oxford OX4 2DQ, UKBlackwell Publishing Asia Pty Ltd, 550 Swanston Street, Carlton, Victoria 3053, Australia

The right of the Authors to be identified as the Authors of this Work has been asserted inaccordance with the Copyright, Designs and Patents Act 1988.

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, recording orotherwise, except as permitted by the UK Copyright, Designs and Patents Act 1988, without theprior permission of the publisher.

First published 2005

Library of Congress Cataloging-in-Publication DataThe endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol.

p. ; cm. a (The encyclopaedia of sports medicine ; v. 11)Includes bibliographical references and index.ISBN-13: 978-1-4051-3017-2 (alk. paper)ISBN-10: 1-4051-3017-2 (alk. paper)1. Endocrine glands. 2. ExerciseaPhysiological aspects. 3. SportsaPhysiological aspects.

[DNLM: 1. Exerciseaphysiology. 2. Endocrine Systemaphysiology. 3. Sportsa

physiology. QT 260 E556 2005] I. Kraemer, William J., 1953– II. Rogol, Alan David, 1941–III. Series.

QP187.3.E93E53 2005612.4—dc22 2004026395

ISBN-13: 978-1-4051-3017-2ISBN-10: 1-4051-3017-2

A catalogue record for this title is available from the British Library

Set in 9/12pt Palatino by Graphicraft Limited, Hong KongPrinted and bound in India by Gopsons Papers Ltd, Noida

Commissioning Editor: Andy RobinsonProject Manager: Alice NelsonDevelopment Editor: Nick MorganProduction Controller: Kate Charman

For further information on Blackwell Publishing, visit our website:http://www.blackwellpublishing.com

The publisher’s policy is to use permanent paper from mills that operate a sustainable forestrypolicy, and which has been manufactured from pulp processed using acid-free and elementarychlorine-free practices. Furthermore, the publisher ensures that the text paper and cover boardused have met acceptable environmental accreditation standards.

Page 7: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

Contents

7 Growth Hormone Variants and HumanExercise, 77wesley c. hymer, richard e.

grindeland, bradley c. nindl and

william j . kraemer

8 Growth Hormone Binding Proteins, 94gerhard baumann

9 Resistance Exercise: Acute and ChronicChanges in Growth Hormone Concentrations,110william j . kraemer, bradley c. nindl

and scott e. gordon

10 The Growth Hormone Response to Acute andChronic Aerobic Exercise, 122arthur l. weltman, laurie wideman,

judy y. weltman and johannes d.

veldhuis

11 Proopiomelanocortin and Exercise, 134heinz w. harbach and

gunter hempelmann

12 Introduction to the Insulin-Like Growth FactorSignaling System, 156charles t. roberts, jr

13 Exercise, Training and the GH–IGF-I Axis, 165alon eliakim, dan nemet and

dan m. cooper

List of Contributors, viii

Foreword, xii

Preface, xiii

1 Introduction, 1alan d. rogol and

william j . kraemer

2 Basic Principles and Mechanisms ofEndocrinology, 8michael r. deschenes and

keiichiro dohi

3 Exercise Testing: a Bridge Between the High-Tech and the Humanathe Need forInnovative Technologies, 25dan m. cooper

4 Measurement of Peptide Hormones, 36martin bidlingmaier, zida wu and

christian j . strasburger

5 Analysis of Low Molecular Weight Substancesin Doping Control, 47mario thevis and wilhelm schänzer

6 The Reproductive Axis, 69johannes d. veldhuis and

arthur l. weltman

v

Page 8: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

vi contents

14 The Role of MGF and Other IGF-I SpliceVariants in Muscle Maintenance andHypertrophy, 180geoffrey goldspink, shi yu yang,

mahjabeen hameed, stephen

harridge and pierre bouloux

15 Adrenal Gland: Fight or Flight Implications for Exercise and Sports, 194michael kjær

16 The Adrenal Medulla: Proenkephalins andExercise Stress, 200jill a. bush and n. travis triplett

17 Exercise and the Hypothalamic–Pituitary–Adrenal Axis, 217warrick j . inder and

gary a. wittert

18 Influence of Energy Availability on Luteinizing Hormone Pulsatility andMenstrual Cyclicity, 232anne b. loucks

19 Oral Contraceptive Use and PhysicalPerformance, 250jaci l. vanheest, carrie e. mahoney

and carol d. rodgers

20 Energy Balance and Exercise-AssociatedMenstrual Cycle Disturbances: Practical andClinical Considerations, 261nancy i . williams and

mary jane de souza

21 Regulation of Testicular Function: Changes in Reproductive Hormones During Exercise, Recovery, Nutritional Deprivation and Illness, 279shalender bhasin

22 Hormonal and Growth Factor-RelatedMechanisms Involved in the Adaptation ofSkeletal Muscle to Exercise, 306fawzi kadi

23 Resistance Exercise and Testosterone, 319atko viru and mehis viru

24 Exercise Response of β-Endorphin and Cortisol: Implications on ImmuneFunction, 339allan h. goldfarb

25 Neuroendocrine Modulation of the ImmuneSystem with Exercise and Muscle Damage, 345mary p. miles

26 The Impact of Exercise on Immunity: the Role of Neuroendocrine–ImmuneCommunications, 368andrea m. mastro and

robert h. bonneau

27 Exercise Regulation of Insulin Action inSkeletal Muscle, 388richard c. ho, oscar alcazar and

laurie j . goodyear

28 Hormone and Exercise-Induced Modulation of Bone Metabolism, 408clifford j . rosen

29 Diet and Hormonal Responses: PotentialImpact on Body Composition, 426jeff s . volek and

matthew j . sharman

30 Neurohumoral Responses and AdaptationsDuring Rest and Exercise at Altitude, 444beth a. beidleman, janet e. staab

and ellen l. glickman

31 Neuroendocrine Influences on TemperatureRegulation in Hot Environments, 466lawrence e. armstrong and

jack a. boulant

32 Alterations in Arginine Vasopressin withExercise, Environmental Stress and OtherModifying Factors, 487carl m. maresh and

daniel a. judelson

Page 9: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

contents vii

33 Human Endocrine Responses toExercise–Cold Stress, 499john w. castellani and

david w. degroot

34 Growth, Maturation and Hormonal Changes During Puberty: Influence of Sport Training, 512james n. roemmich

35 Effects of Testosterone and Related Androgenson Athletic Performance in Men, 525karl e. friedl

36 Growth Hormone and Sport, 544jennifer d. wallace and

ross c. cuneo

37 Endocrinology of Overtraining, 578andrew c. fry, juergen m.

steinacker and romain meeusen

38 Endocrinology of Sport Competition, 600jay r. hoffman

Index, 613

Page 10: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

OSCAR ALCAZAR PhD, Research Division, JoslinDiabetes Center and Department of Medicine, HarvardMedical School, Boston, MA 02215, USA

LAWRENCE E. ARMSTRONG PhD,Departments of Kinesiology and Physiology-Neurobiology,University of Connecticut, Storrs, CT 06269, USA

GERHARD BAUMANN MD, Division ofEndocrinology, Metabolism and Molecular Medicine,Northwestern University Feinberg School of Medicine andVeterans Administration Chicago Health Care System, 303East Chicago Avenue, Chicago, IL 60611, USA

BETH A. BEIDLEMAN DSc, Biophysics andBiomedical Modeling Division, US Army Research Institutefor Environmental Medicine, Natick, MA 01760, USA

SHALENDER BHASIN MD, UCLA School ofMedicine, Drew-UCLA Reproductive Science ResearchCenter, Division of Endocinology, Metabolism andMolecular Medicine, Charles R. Drew University ofMedicine and Science, Los Angeles, CA 90059, USA

MARTIN BIDLINGMAIER MD,Neuroendocrine Unit, Medizinische Klinik—Innenstadt,Klinikum der LMU, Ziemssenstr. 1, 80336 Munich,Germany

ROBERT H. BONNEAU PhD, Department ofMicrobiology and Immunology, Pennsylvania StateUniversity College of Medicine, Milton S. Hershey MedicalCenter, Hershey, PA 17033, USA

JACK A. BOULANT PhD, Department ofPhysiology and Cell Biology, Ohio State University Collegeof Medicine, Columbus, OH 43210, USA

PIERRE BOULOUX MD, Department of Medicine,Royal Free and University College Medical School,University of London, Royal Free Campus, Rowland HillStreet, London, NW3 2PF, UK

JILL A. BUSH PhD, Laboratory of IntegratedPhysiology, Department of Health and Human Performance,University of Houston, Houston, TX 77204, USA

JOHN W. CASTELLANI PhD, Thermal andMountain Medicine Division, US Army Research Institutefor Environmental Medicine, 42 Kansas Street, Natick, MA01760-5007, USA

DAN M. COOPER MD, Center for the Study ofHealth Effects of Exercise in Children, Department of Pediatrics, Irvine College of Medicine, University ofCalifornia, Irvine, CA 92868, USA

ROSS C. CUNEO PhD, Department of Diabetes and Endocrinology, University of Queensland, PrincessAlexandra Hospital, 4102 Brisbane, Queensland, Australia

DAVID W. DEGROOT MS, Thermal andMountain Medicine Division, US Army Research Institutefor Environmental Medicine, 42 Kansas Street, Natick, MA01760-5007, USA

MICHAEL R. DESCHENES PhD, Departmentof Kinesiology, The College of William and Mary,Williamsburg, VA 23187-8795, USA

MARY JANE DE SOUZA PhD, Women’sExercise and Bone Health Laboratory, Faculty of PhysicalEducation and Health, 52 Harbord Street, University ofToronto, Toronto, Ontario, M5S 2W6, Canada

List of Contributors

viii

Page 11: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

list of contributors ix

KEIICHIRO DOHI PhD, Osaka University ofHealth and Sport Sciences, Asashirodai, Kumatori-cho,Sennan-gun, Osaka, 590-0496, Japan

ALON ELIAKIM MD, Sackler School of Medicine,Tel-Aviv University and Child Health and Sports Center,Pediatric Department, Meir General Hospital, Kfar-Saba44281, Israel

KARL E. FRIEDL PhD, US Army Research Instituteof Environmental Medicine, Natick, MA 01760-7007, USA

ANDREW C. FRY PhD, Exercise BiochemistryLaboratory, 135 Roane Field House, The University ofMemphis, Memphis, TN 38152, USA

ELLEN L. GLICKMAN PhD, School of Exercise,Leisure and Sport, Kent State University, Kent, OH 44513,USA

ALLAN H. GOLDFARB PhD, Exercise and Sport Science Department, University of North Carolina-Greensboro, Greensboro, NC 27402-6170, USA

GEOFFREY GOLDSPINK PhD, ScD,Department of Surgery, Royal Free and University CollegeMedical School, University of London, Royal Free Campus,Rowland Hill Street, London, NW3 2PF, UK

LAURIE J . GOODYEAR PhD, Joslin DiabetesCenter, One Joslin Place, Boston, MA 02215, USA

SCOTT E. GORDON PhD, Human PerformanceLaboratory, East Carolina University, Greenville, NC 27858,USA

RICHARD E. GRINDELAND PhD, LifeScience Division, NASA-Ames Research Center, MoffettField, CA 94035, USA

MAHJABEEN HAMEED PhD, Department ofSurgery, Royal Free and University College Medical School,University of London, Royal Free Campus, Rowland HillStreet, London, NW3 2PF, UK

HEINZ W. HARBACH MD, Department ofAnesthesiology, Intensive Care Medicine, Pain Therapy,University Hospital, Giessen, Rudolf-Buchheim-Str. 7, D-35385 Giessen, Germany

STEPHEN HARRIDGE PhD, Department ofPhysiology, Royal Free and University College Medical

School, University of London, Royal Free Campus, RowlandHill Street, London, NW3 2PF, UK

GUNTER HEMPELMANN MD, Department of Anesthesiology, Intensive Care Medicine, Pain Therapy,University Hospital, Giessen, Rudolf-Buchheim-Str. 7, D-35385 Giessen, Germany

RICHARD C. HO PhD, Research Division, JoslinDiabetes Center and Department of Medicine, HarvardMedical School, Boston, MA 02215, USA

JAY R. HOFFMAN PhD, Department of Health andExercise Science, College of New Jersey, Ewing, NJ 08628,USA

WESLEY C. HYMER PhD, Department ofBiochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA

WARRICK J . INDER MD, Department ofMedicine, St. Vincent’s Hospital, University of Melbourne,Fitzroy, VIC 3065, Australia

DANIEL A. JUDELSON MA, HumanPerformance Laboratory, Department of Kinesiology,University of Connecticut, Storrs, CT 06269-1110, USA

FAWZI KADI PhD, Department of Physical Educationand Health, Örebro University, 70182 Örebro, Sweden

MICHAEL KJÆR MD, PhD, University ofCopenhagen, Sports Medicine Research Unit, BispebjergHospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV,Denmark

WILLIAM J. KRAEMER PhD, HumanPerformance Laboratory, Department of Kinesiology,University of Connecticut, Storrs, CT 06269-1110, USA

ANNE B. LOUCKS PhD, Department of BiologicalSciences, Ohio University, Irvine Hall 053, Athens, OH45701, USA

CARRIE E. MAHONEY BS, Department ofKinesiology, University of Connecticut, Storrs, CT 06269-1110, USA

CARL M. MARESH PhD, Human PerformanceLaboratory, Department of Kinesiology, University ofConnecticut, Storrs, CT 06269-1110, USA

Page 12: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

x list of contributors

ANDREA M. MASTRO PhD, Department ofBiochemistry and Molecular Biology, 431 South FrearBuilding, Pennsylvania State University, University Park,PA 16802, USA

ROMAIN MEEUSEN PhD, Faculty of PhysicalEducation and Physiotherapy, Vrije Universiteit Brussel,Brussels, 1050, Belguim

MARY P. MILES PhD, Department of Health andHuman Development, Montana State University, Bozeman,MT 59717, USA

DAN NEMET MD, Sackler School of Medicine, Tel-Aviv University and Child Health and Sports Center,Pediatric Department, Meir General Hospital, Kfar-Saba44281, Israel

BRADLEY C. NINDL PhD, Military PerformanceDivision, US Army Research Institute of EnvironmentalMedicine, Natick, MA 01760, USA

CHARLES T. ROBERTS, JR PhD, Departmentof Pediatrics (NRC5), Oregon Health and ScienceUniversity, 3181 SW Sam Jackson Park Rd., Portland, OR97239, USA

CAROL D. RODGERS PhD, Department ofPhysical Education and Health, University of Toronto,Toronto, Ontario, Canada, and Department of Physiology,Faculty of Medicine, University of Toronto, Ontario, M5S2W6, Canada

JAMES N. ROEMMICH PhD, Department ofPediatrics, Division of Behavioral Medicine, State Universityof New York at Buffalo, 3435 Main Street, Buffalo, NY14214-3000, USA

ALAN D. ROGOL MD, PhD, Clinical Pediatrics,University of Virginia, ODR Consulting, 685 ExplorersRoad, Charlottesville, VA 22911-8441, USA

CLIFFORD J . ROSEN MD, Maine Center forOsteoporosis Research and Education, St. Joseph Hospital,900 Broadway, Bangor, ME 04401, USA

WILHELM SCHÄNZER PhD, Institute ofBiochemistry, German Sport University Cologne, Carl-DiemWeg 6, 50933 Cologne, Germany

MATTHEW J. SHARMAN MA, HumanPerformance Laboratory, Department of Kinesiology, 2095

Hillside Road, Unit 110, University of Connecticut, Storrs,CT 06269-1110, USA

JANET E. STAAB BS, Thermal and MountainMedicine Division, US Army Research Institute forEnvironmental Medicine, 42 Kansas Street, Natick, MA01760-5007, USA

CHRISTIAN J . STRASBURGER MD,Division of Endocrinology, Department of Internal Medicine,Charité—Campus Mitte, Schumannstrasse 20/21, 10117Berlin, Germany

JUERGEN M. STEINACKER MD, PhD,Section of Sports and Rehabilitation Medicine, University ofUlm, 89070 Ulm, Germany

MARIO THEVIS PhD, Institute of Biochemistry,German Sport University Cologne, Carl-Diem Weg 6, 50933Cologne, Germany

N. TRAVIS TRIPLETT PhD, Department ofHealth, Leisure and Exercise Science, Appalachian StateUniversity, Boone, NC 28608, USA

JACI L. VANHEEST PhD, Department ofKinesiology, University of Connecticut, Storrs, CT 06269,USA, and Adjunct in Department of Physical Education andHealth, University of Toronto, Toronto, Ontario, M5S 2W6,Canada

JOHANNES D. VELDHUIS MD, Division ofEndocrinology and Metabolism, Department of InternalMedicine, Mayo Medical and Graduate Schools of Medicine,General Clinical Research Center, Mayo Clinic, Rochester,MN 55905, USA

ATKO VIRU DSc, PhD, Institute of Sports Biology,University of Tartu, 18 Ylikooli, Tartu 51014, Estonia

MEHIS VIRU PhD, Institute of Sports Biology,University of Tartu, 18 Ylikooli, Tartu 51014, Estonia

JEFF S. VOLEK PhD, Department of Kinesiology,University of Connecticut, Storrs, CT 06269-1110, USA

JENNIFER D. WALLACE PhD (Med)pending, Metabolic Research Unit, Department of Medicine,University of Queensland, Princess Alexandra Hospital,4102 Brisbane, Queensland, Australia

Page 13: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

list of contributors xi

ARTHUR L. WELTMAN PhD, Department ofHuman Services, Department of Medicine, General ClinicalResearch Center and Exercise Physiology Laboratory,Memorial Gymnasium, University of Virginia,Charlottesville, VA 22908, USA

JUDY Y. WELTMAN MS, General ClinicalResearch Center, University of Virginia, Charlottesville, VA 22908, USA

LAURIE WIDEMAN PhD, Department of Exerciseand Sport Science, University of North Carolina-Greensboro,Greensboro, NC 27402-6170, USA

NANCY I . WILLIAMS ScD, Noll PhysiologicalResearch Center and Department of Kinesiology, 108 Noll

Laboratory, Penn State University, University Park, PA16802, USA

GARY A. WITTERT MD, Department of Medicine,Royal Adelaide Hospital, University of Adelaide, Adelaide,SA 5000, Australia

ZIDA WU MD, Division of Endocrinology, Departmentof Internal Medicine, Charité—Campus Mitte,Schumannstrasse 20/21, 10117 Berlin, Germany

SHI YU YANG PhD, MVet Sci, Department ofSurgery, Royal Free and University College Medical School,University of London, Royal Free Campus, Rowland HillStreet, London, NW3 2PF, UK

Page 14: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

The Encyclopaedia of Sports Medicine is a highlyvaluable series that has been produced during thelast 17 years by the IOC Medical Commission. Thepresent edition becomes the eleventh volume in theseries.

A volume that is focused on the physiology ofsport and exercise could be expected to concentrateon skeletal muscle metabolism and the supportingfunctions of the cardiovascular and respiratory sys-tems. Indeed, these aspects of the biology of humanperformance have been featured in certain of the earl-ier editions of The Encyclopaedia of Sports Medicine.

It is now entirely appropriate that attention begiven to the ductless glands that produce the hor-mones that modify and control so many of theimportant body functions. A great number of the

chronic adaptations of the human organism to sportconditioning result in major changes in endocrinefunction. The co-editors and contributing authors of Encyclopaedia Vol. XI, The Endocrine System inSports and Exercise, have provided a comprehensiveand authoritative coverage of this highly complexsystem. This volume will serve as a leading referencefor physicians, scientists and graduate students formany years to come.

I am pleased to congratulate the co-editors andthe contributing authors on the quality of their workand to welcome the newest volume into the presti-gious Encyclopaedia of Sports Medicine series.

Dr Jacques RoggeIOC President

Foreword

xii

Page 15: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

It is an honor for each of us to serve as Co-Editors for this important contribution in the field of endo-crinology, more specifically sport and exerciseendocrinology. We were fortunate to have an excep-tional group of scientists to participate in this sem-inal work in our field. Thus, each chapter is writtenby one or more of the world’s leading scientists intheir specific area of expertise. Their enthusiasmand excitement for the project and its importantance is reflected in the very dynamic of each chapter. Weacknowledge many of our other distinguished col-leagues who have also made influential contribu-tions to this field but were unable to participate inthis project, as this field of study has undergonedramatic growth over the past 20 years.

Each author was asked to develop a workingparadigm that would not only provide a cuttingedge overview of the field as it currently exists butwould also provide a template for research over thecoming years. This encyclopedia provides one of the few resources for such a comprehensive view of the many aspects of endocrinology in sport andexercise. It is important to understand that eachchapter was not meant to be a comprehensive reviewof the existing literature, but rather a state-of-the-artconceptual framework for the area covered. Thus, itwas not the purpose to provide exhaustive refer-ences but rather a “cutting edge” perspective for useby both clinical and basic scientists. It is our hopethat this encyclopedia serves to educate as well asinspire future research in the area of endocrinologyin sport and exercise.

For so many years the field of exercise endocrino-logy has been folded into the many other areas of

physiology, lacking direct acknowledgment of itsown value as a discipline. The study of endocrino-logy as a specialty in medicine has been in existencefor decades, while its applications in the field ofexercise and sport has been a more recent phe-nomenon with typical focus on one or, at most, a fewhormones. This encyclopedia provides the readerwith a more complete view of the many targetedareas of study that have propelled this field for-ward over the past several decades. The number ofpublications listed on PubMed with hormones andexercise has increased by more than 30-fold over thepast 20 years. Advances in technology as well asgreater interest in the endocrine system and its integration with the nervous and immune systemshave fueled the exponential rise in publications inendocrinology and exercise.

The encyclopedia begins with a basic overview ofthe principles and concepts in the field of endo-crinology. It then takes the reader through the different endocrine glands allowing multiple per-spectives on function. The interactive effects of hormonal influences on the immune system, muscleand bone are then examined. The interactions withnutrition are then covered, providing a unique viewof one of the most important (and controversial)aspects of sport and exercise performance. The textthen provides a unique examination of the hor-monal interfaces with environmental stresses. Thestudy of the underlying hormonal aspects of sportitself remains a much less developed area of studydue to the inability to access athletes under com-petitive stress in game/match/meet conditions. Inthis encyclopedia, chapters examining overtraining,

Preface

xiii

Page 16: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the young athlete’s growth and development, andthe endocrinology of sport competition are covered,providing some unique perspectives for futurework. The problem of anabolic drug use is thenexamined in detail. Ending with the study of endocrine mechanisms involved with competitivestress, this volume of The Encyclopaedia of SportsMedicine series permits the student, clinician andpracticing scientist the chance to view this fieldmore comprehensively. We are particularly indebted

to the International Olympic Committee’s MedicalCommission and Sub-Commission on Publicationsin the Sport Sciences for providing us this opportun-ity to present to the world our field of study in itsfull array.

William J. KraemerStorrs, Connecticut

Alan D. RogolCharlottesville, Virginia

xiv preface

Page 17: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

1

What is endocrinology?

Endocrinology is the science of intercellular andintracellular communication. Classically, as definedby Bayliss and Starling more than 100 years ago, itwas the secretion of a chemical substance (hormone,from the Greek word hormaein, to stimulate, to spurinto action) into the general circulation to cause an effect at a distal site (target organ). Hormonereceptors were unknown at that time. The hypo-thesis as stated was used to describe the secretionand action of secretin, a chemical substance secretedby the small intestine into the blood stream to stimu-late pancreatic exocrine secretion (Bayliss & Starling1902).

Initially all hormones were considered producedin specialized glandsafor example, the thyroid, theadrenals or the pituitaryato act on single or mul-tiple organs as a means of homeostatic regulation of metabolism (biochemical control mechanisms)agene expression; biosynthetic pathways and theirenzymatic catalysis; the modification, transforma-tion and degradation of biological substances; thebiochemical mediation of the actions and interactionsof such substances; and the means for obtaining,storing and mobilizing stored energy (Table 1.1).This fundamental concept has been subsequently(and greatly) expanded to account for many formsof intercellular (and even intracellular) transfer ofinformation in plants and animals (Table 1.2). Hor-mones represent many classes of biological mole-cules (Table 1.3). It now includes almost every tissueand cell for they either produce or respond to hor-mones. These hormonal systems integrate informa-

tion in conjunction with the neural and immune systems to transmit it to define ambient conditionsand to maintain metabolic homeostasis. These fun-damental concepts have also been greatly expandedby advances in cell biology, molecular biology andgenetics to help explain hormonal synthesis, actionand perhaps integration, but the backbone of theendocrine system remains unchangedaintegrationof multiple systems and the maintenance of cellularand metabolic homeostasis.

Classically, hormones were defined by an ablation–replacement paradigm. Endocrine glandswere removed from normal animals, the glandground-up and extracted for the active substance.Restoration, by the extract of the function lost byremoval of the gland, closed the loop and led to thediscovery of many active hormonal compounds.Over 150 years ago, Berthold showed the ‘proof-of-concept’ of hormones by castrating roosters andnoting regrowth of the wattles and comb by replace-ment of the testis into a body cavity (Berthold 1849).The transplant was both ectopic and lacked innerva-tion, permitting him to conclude that the testesreleased a product that controlled the development(and maintenance) of the secondary sexual charac-teristics. The concept of homeostasis was stated byClaude Bernard, who showed that the liver couldrelease glucose into the blood:

The constancy of the internal environment is the condition that life should be free andindependent. . . . So far from the higher animalbeing indifferent to the external world, it is onthe contrary in a precise and informed relation

Chapter 1

Introduction

ALAN. D. ROGOL AND WILLIAM J. KRAEMER

Page 18: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

2 chapter 1

Table 1.1 Selected hormones of the endocrine system and their actions.

Endocrine organ Hormone Major actions

Testes Testosterone Stimulates development and maintenance of male sex characteristics, growth and protein anabolism

Ovaries Estrogens Develops female secondary sex characteristics; matures theepiphyses of the long bones

Progesterone Develops female sex characteristics; maintains pregnancy; develops mammary glands

Anterior pituitary Growth hormone Stimulates IGF-I and -II synthesis; stimulates protein synthesis, growth and intermediary metabolism

Adrenocorticotropic hormone (ACTH) Stimulates glucocorticoid release in adrenal cortexThyroid-stimulating hormone (TSH) Stimulates thyroid hormone synthesis and secretionFollicle-stimulating hormone (FSH) Stimulates growth of follicles in ovary, seminiferous

tubules in testes and sperm productionLuteinizing hormone (LH) Stimulates ovulation and production and secretion of sex

hormones in ovaries and testesProlactin (Prl) Stimulates milk production in mammary glands

Posterior pituitary Antidiuretic hormone (ADH) Increases reabsorption of water by kidneys and stimulates contraction of smooth muscle

Oxytocin Stimulates uterine contractions and release of milk by mammary glands

Adrenal cortex Glucocorticoids Inhibits or retards amino acid incorporation into proteins (cortisol); stimulates conversion of proteins into carbohydrates (gluconeogenesis); maintains normal blood sugar level; conserves glucose; promotes metabolism of fat

Mineralcorticoids Increases or decreases sodium–potassium metabolism; (aldosterone, increases body waterdeoxycorticosterone, etc.)

Adrenal medulla Epinephrine Increases cardiac output; increases blood sugar, glycogen breakdown and fat mobilization

Norepinephrine (some) Similar to epinephrine plus constriction of blood vesselsProenkephalins (e.g. peptide F, E) Analgesia; enhances immune function

Thyroid Thyroxine Stimulates oxidative metabolism in mitochondria and cell growth

Calcitonin Reduces blood calcium levels; inhibits osteoclast function

Heart (cardiocytes) Atrial natriuretic hormone Facilitates the excretion of sodium and water; regulates blood pressure and volume homeostasis and opposes the actions of the renin–angiotensin system

Pancreas Insulin Stimulates absorption of glucose and storage as glycogenGlucagon Increases blood glucose levels

Parathyroids Parathyroid hormone Increases blood calcium; decreases blood phosphate

Skin Vitamin D Produces vitamin D from 7-dehydrocholesterol and sunlight

Adipose tissue Leptin Regulates appetite and energy expenditure

IGF-I, insulin-like growth factor I; IGF-II, insulin-like growth factor II.

Page 19: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

introduction 3

with it, in such a way that its equilibriumresults from a continuous and delicate compen-sation, established as by the most sensitive ofbalances (Bernard 1957).

Both of these results were known to Bayliss andStarling when they stated the secretin hypothesisand coined the word hormone. Later, Walter Cannonemphasized that the constancy of the internal envir-onment could only be achieved through the opera-tion and integration of exquisitely co-ordinatedphysiologic processes which he named homeostatic(Cannon 1939).

Hormones may be fully active when secreted, for example cortisol; however, some may requiremodification to become fully active, for example theconversion of tetraiodothyronine (thyroxine, T4) totriiodothyronine (T3) by a specific deiodinase, theconversion of testosterone to dihydrotestosterone(DHT) by 5-α reductase and the conversion of vit-amin D3 to 1,25 dihydroxyvitamin D by two distincthydroxylases. Post-translational modification byphosphorylation, sulfation, or the addition of a lipidchain may be required to permit solubility in theparticular environment that the hormone finds itself.

Because hormones may work at a distance, noveltransport systems have evolved. Some steroid hormones have specific binding proteins to permitsolubility in aqueous environments (sex steroidsbind to sex hormone binding globulin [SHBG] andto albumin). There is a large series of insulin-like

growth factor binding proteins (IGFBPs) that carryIGF-I and II to their sites of action, prolong their circulating half-life and dampen their effects ashypoglycemic agents (LeRoith 2003).

Most hormones are not secreted at a constant rate.Thus, the circulating concentrations undergo fluctu-ations that may be due to intermittent secretion oralterations in metabolism (clearance). For example,the circulating concentrations of insulin are not uni-form, but vary from minute-to-minute according tothe timing and composition of meals; from a base-line that is often determined by the relative mass oflean and fat tissue and the regional distribution ofthe fat tissue (visceral versus subcutaneous). Theconcentrations of growth hormone vary in ‘pulses’throughout the day in response to meals and tosleep. Those of the gonadotropins, luteinizing hor-mone (LH) and follicle-stimulating hormone (FSH)and the sex steroids, estradiol and progesterone,vary over the approximately 28 days of the men-strual cycle in addition to the 1- to 2-hourly pulses of LH during the day. Adrenocorticotropic hor-mone (ACTH) and cortisol usually maintain a dailyrhythm (high early in the morning and low at night)in those who sleep nocturnally. Disruption of theserhythms can lead to infertility or jet-lag.

What do hormones do?

For signal recognition the target cell produces ahighly specific ‘receptor’ that recognizes the chemical

Table 1.2 Cellular communication.

Endocrine Chemical messenger (hormone) formed in a specialized tissue or organ (gland) and released into a circulation to affect another organ at a distance

Paracrine Hormone synthesized in specialized cells to act on nearby cellsJuxtacrine Hormone synthesized in one cell and acts on a contiguous cellAutocrine Hormone synthesized in one cell and acts on the surface of the same cellIntracrine Hormone synthesized in one cell and acts on the internal machinery of that cell without the necessity

of cell surface receptors

Amino acid derivative Epinephrine, thyroid hormonesSteroid Testosterone, cortisol, vitamin DPeptide Gonadotropin-releasing hormone, secretinFatty acid derivatives Prostaglandins, leukotrienes

Table 1.3 Chemical classes ofhormonal molecules.

Page 20: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

4 chapter 1

message (primary messenger) and begins a cascadeof chemical reactions that lead to an alteration in theoutput of that cell, for example protein synthesis.Receptor proteins may be expressed at the cell sur-face, cytoplasm and the nucleus. The receptorsthemselves may be part of the overall regulatingmechanism, for the signals from the hormones maybe modified by their number and affinity for theirligand. Receptor endocytosis leads to the inter-nalization of cell surface receptors; the hormone–receptor complex is subsequently dissociated andleads to a diminution of hormonal effect. Receptordesensitization (attenuated receptor signaling in thepresence of ligand) may alter the receptor’s affinityfor the ligand, for example by phosphorylation (forepinephrine). The major hormonal signaling path-ways use G-protein coupled receptors, tyrosinekinase receptors, serine/threonine kinase receptors,ion channels, cytokine receptors and nuclear recep-tors to activate the intracellular machinery.

Control of hormone secretion

The endocrine system is organized in a hierarchalmanner. As an example, the hypothalamus releasesthyrotropin-releasing hormone (TRH) which stimu-lates the anterior pituitary to produce thyroid-stimulating hormone (TSH) which, in turn, acts onthe thyroid to increase the synthesis and release ofthe thyroid hormones, T4 and T3. The thyroid hor-mones circulate bound to a specific binding protein,thyroxine binding protein (TBG), to affect virtuallyall cells in the body through intracellular thyroidhormone receptors and usually regulate the rate ofmetabolism of their targets (Fig. 1.1).

All of these systems are regulated by a series ofnegative (and some positive) feedback loops tomaintain the system within a narrow range of oper-ation (homeostasis). However, this complex, hierar-chal system does not operate in a vacuum. There aremultiple points of contact of the many endocrineaxes with the neural and immune systems, as ex-emplified by the organism’s response to stress (Fig. 1.2) (Selye 1950), which is served by a complexinteraction of the central nervous system (CNS) andperipheral organs. This system must receive (andintegrate) multiple neurosensory signals, for ex-

ample visual and visceral inputs, blood borne andlimbic signals. The concerted activation of this sys-tem leads to multiple outputs including physicaland behavioral changes. When these are adaptive,one usually remains within the homeostatic range(see above). The hierarchy of this response isinstructive for many of the other interactions of theneural, endocrine and immune systems.

The central components include the parvocellularcorticotropin-releasing hormone (CRH) and argininevasopressin (AVP) neurons within the paraventricu-lar nucleus of the hypothalamus, the CRH neuronsof the paragigantocellular and parabranchial nucleiof the medulla and the locus ceruleus (LC), andother mostly noradrenergic (norepinephrine [NE])cell groups of the medulla and pons (the LC/NEsympathetic system, see Fig. 1.2). The peripherallimbs are the hypothalamic–pituitary–adrenal (HPA)axis, the efferent sympathetic–adrenomedullary sys-tem and some components of the parasympatheticnervous system. AVP and CRH activate the pituit-ary to produce proopiomelanocortin (POMC), whichis eventually cleaved to produce ACTH (and manyother centrally active peptides, such as α-melano-cyte stimulating hormone, α-MSH). ACTH activates

CNS inputs

Pituitary

Pituitary trophichormone

Tier IHypothalamichormones

Tier IIParacrinecytokines andgrowth factors

Tier IIIPeripheralhormones

Hypothalamus

Target gland

Fig. 1.1 Model for regulation of anterior pituitaryhormones secretion by three tiers of control.Hypothalamic hormones impinge directly on theirrespective target cells. Intrapituitary cytokines andgrowth factors regulate tropic cell function by paracrine(and autocrine) control. Peripheral hormones exertnegative feedback inhibition of respective pituitarytrophic hormone synthesis and secretion. (Reproducedwith permission from Ray & Melmed 1997.)

Page 21: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

introduction 5

the adrenal glands to produce cortisol and othersteroids that affect metabolic homeostasis and theimmune system. There are further interactions withthe gonad axis (inhibition at multiple levels), thehypothalamic–pituitary–growth hormone/IGF-I axis(inhibition at multiple levels) and the thyroid axis(decreased production of TSH and inhibition of theconversion of T4 to the more metabolically activeT3ain fact it has been called the ‘euthyroid sick’ syn-drome in which there are diminished amounts of

TSH and the peripheral thyroid hormones are lessactive). Further multiple metabolic axes are affectedby antagonizing the effects of growth hormone andsex steroids on fat tissue catabolism and muscle andbone anabolism (Fig. 1.3).

Exercise and endocrinology

Exercise produces dramatic challenges to the home-ostatic mechanisms. The acute exercise responsescan see metabolic increases of 10-fold or more. Forceproduction can be repetitively high and maximallimits reached in typical training sessions. The challenges to the body under conditions of athleticcompetition are also dramaticaranging from per-formance of a marathon race in under 2 h and 10 minto an Olympic weightlifter lifting a weight four timeshis body mass. The mechanisms by which exerciseis tolerated and adapted to are intimately related tohormonal regulation of physiological systems, butwith both acute and chronic changes.

Over the past 50 years or more, exercise and sportphysiology has continued to increase its study ofhormonal mechanisms that mediate the exercise-induced adaptations. For example, in resistance

Circadianrhythms Stress

Behavioral adaptation

Arcuate NPOMC

Analgesia

NPY

CRHLC/NE

Symp. Syst.

GABA/BZD

SerotoninAcetylcholine

SPAVP

ACTH

EpinephrineNorepinephrineGlucocorticoids

Peripheral adaptation

CRH

NE

Fig. 1.2 Simplified representation of the central andperipheral components of the stress system, theirfunctional interrelations and their relationships to othercentral nervous systems involved in the stress response.Activation is represented by solid lines and inhibition by dashed lines. ACTH, adrenocorticotropic hormone;AVP, arginine vasopressin; BZD, benzodiazepine; CRH,corticotropin-releasing hormone; GABA, γ-aminobutyricacid; LC/NE Sym. Syst., locus ceruleus/norepinephrine–sympathetic system; NE, norepinephrine; NPY,neuropeptide Y; POMC, proopiomelanocortin; SP,substance P. (Adapted with permission from Chrousos & Gold 1992. Copyright 1992, American MedicalAssociation.)

StressSS GnRH

LH, FSH

Visceraladiposity

Bonemass

Sexsteroids

GlucocorticoidsGH

Fig. 1.3 Schematic representation of the detrimentaleffects of chronic stress on adipose tissue metabolism and bone mass. Stimulation is represented by solid linesand inhibition by light lines. FSH, follicle-stimulatinghormone; GH, growth hormone; GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone; SS,somatostatin. (Adapted with permission from Tsigos & Chrousos 1995.)

Page 22: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

6 chapter 1

training of primary importance to acute exerciseperformance and subsequent tissue remodeling isthe role(s) played by many members of the endo-crine system (Kraemer & Ratamess 2003). Hormonalelevations in response to resistance exercise takeplace in a unique physiological environment. Acuteelevations in circulating blood hormone concentra-tions (i.e. resulting from either increased secretion,reduced hepatic clearance, plasma volume reduc-tions, reduced degradation rates) observed bothduring and immediately following a resistanceexercise protocol present a greater likelihood ofinteraction with receptors on either the target tissuecell membrane (e.g. peptides) or with nuclear/cyto-plasmic receptors located within the target tissue(e.g. steroid receptors) (Kraemer 2000). Coincidingwith blood hormonal concentrations is the numberof available receptors for binding and subsequentcellular changes. Interaction with the receptor ini-tiates a myriad of events culminating in a specificresponse, such as an increase in muscle protein synthesis. Therefore, from the role of anabolic hor-mones (e.g. growth hormone, testosterone, IGFs) inprotein synthesis in response to resistance train-ing to insulin’s role in glycogen metabolism withendurance training, hormonal mechanisms havebecome prominent in the study of exercise andsport. Due to the ubiquitous nature of hormones, no physiological system can adequately functionwithout one or more involved with its response andadaptation to one form or another of exercise. Suchdramatic hormonal influences have increased theinterest in endocrinology for those who investigateexercise and sport.

With exercise creating such a unique physiolo-gical environment, one cannot merely extrapolateour understanding of resting homeostatic physiology(endocrinology). The conditions with exercise are

such that the stimulus is highly specific in its nature.Different from the general nature of stress proposedby Selye (1950) over 50 years ago, we now know thatstress is very specific in its ‘fingerprint’ and mediat-ing mechanisms. Thus, the magnitude of hormonalresponses as well as the biocompartments in whichthey occur can differ dramatically. For example,with a resistance exercise that just stimulates an armmuscle, one notes only small or no increases in circulating anabolic hormones, but growth factorconcentrations (for example, IGF-I) may dramatic-ally increase, specifically at the site of their action.Differences in hormonal responses also occur withthe magnitude of the exercise intensityalow in-tensity exercise produces a smaller magnitude ofhormonal response than higher intensity exercise.Thus, the influence of work, intensity, volume andfrequency all help create the exercise stimulus thatoccurs acutely with a single exercise session orrepetitively as one continues with training.

Understanding the role of different hormoneswithin and among the various physiological com-munication systems is a challenge as few, if any,hormones act in isolation. Furthermore, with mul-tiple-level communication being so important tooptimal homeostatic regulation, complex integra-tion of hormonal signals is required to respond tothe multiple energy demands of exercise.

Finally, the study of hormones and their roles inexercise and sport has permitted a better under-standing of the stresses of competition, overtrainingand identified key factors in exercise prescription(e.g. intensities, frequencies and durations) that can be optimized to create improved training pro-grams and, ultimately, performance. Ultimately, theunderlying physiological basis of any exercise orsport stress has its underpinnings in endocrinolo-gical science.

References

Bayliss, V.M. & Starling, E.H. (1902) Themechanism of pancreatic secretion.Journal of Physiology 28, 325–334.

Bernard, C. (1957) An Introduction to theStudy of Experimental Medicine. Dover,New York.

Berthold, A.A. (1849) Transplantation der

hoden. Arch Anat Physiol Wiss Med 16,42–46.

Cannon, W.B. (1939) The Wisdom of theBody. Norton, New York.

Chrousos, G.P. & Gold, P.W. (1992) Theconcepts of stress and stress systemdisorders. JAMA 267, 1244–1252.

Kraemer, W.J. (2000) Neuroendocrineresponses to resistance exercise. In:Essentials of Strength Training andConditioning, 2nd edn. (Baechle, T., ed.).Human Kinetics, Champaign, IL:91–114.

Kraemer, W.J. & Ratamess, N.A. (2003)

Page 23: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

introduction 7

Endocrine responses and adaptations tostrength and power training. In: Strengthand Power in Sport, 2nd edn. (Komi, P.V.,ed.). Blackwell Science, Malden, MA:361–386.

Le Roith, D. (2003) The insulin-like growthfactor system. Experimental DiabesityResearch 4, 205–212.

Tsigos, C. & Chrousos, G.H. (1995) Stress,endocrine manifestations and diseases.In: Handbook of Stress Medicine (Cooper,G.L., ed.). CRC Press, Boca Raton, FL:61–65.

Ray, D. & Melmed, S. (1997) Pituitarycytokine and growth factor expressionand action. Endocrine Reviews 18,206–228.

Selye, H. (1950) Stress and the generaladaptation syndrome. British MedicalJournal 4667, 1383–1392.

Page 24: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

8

Introduction and general principles

The overarching theme of physiology is the per-sistent effort to maintain homeostasis within theorganism. This drive to sustain a constant internalmilieu is challenged by regular perturbations of theorganism’s internal and external environment. Inthe face of these changes, the capacity to achievehomoeostasis is determined by effective commun-ication among the cells of the organism. Two sys-tems have evolved to provide such communication.The nervous system generally involves immediateand brief responses to stimuli. In contrast, theendocrine system typically is slower to respond toalterations in the environment, but its responsespersist longer than those of the nervous system. Theeffects of the endocrine system are pervasive, andregulate the activity of virtually all cells within thebody. Each of these cells is perfused with blood; theendocrine system capitalizes on this physiologicalarrangement to communicate effectively through-out the body.

The word ‘hormone’ derives from the Greekword hormaein which means ‘to spur into action’ or‘to stimulate’. In 1902, Bayliss and Starling (1902)described a substance secreted into the blood by oneorgan (small intestine) that gave rise to a response inanother (pancreas). Thus, secretin became the firsthormone identified. Today, a hormone is commonlydefined as ‘a chemical substance that is released intothe blood in small amounts and that, after deliveryby the circulation, elicits a typical physiologicalresponse in other cells’ (Goodman 1994). It has been discovered, however, that in addition to this

example of classical endocrine function, mediatingagents can be released into the interstitial fluid todiffuse to and affect neighboring cells (paracrineeffect), or even to interact with the same cell thatsecreted them (autocrine effect). Indeed, some sub-stances, for instance insulin-like growth factor I(IGF-I), may exert their biological responses viaendocrine, paracrine and autocrine routes (Yakar et al. 2002). More recently it has been proposed (Re2003) that some growth factors and peptide hor-mones may directly regulate activity within the cellof synthesis without ever exiting that cell (intracrineeffect). For the purpose of this chapter, only the fea-tures of the endocrine system will be addressed.

Although scores of hormones have been identi-fied, and their biological activities regulate a plethoraof physiological processes, several fundamentalprinciples of endocrine function have been estab-lished. First, hormones are synthesized and secretedby specialized ductless endocrine glands into thebloodstream to be carried to ‘target’ cells that bindthe hormone and respond to it by altering their biological activity in a specific, preprogrammedfashion. Second, although some endocrine glandsare the main components of organs specialized forendocrine function (e.g. pituitary, thyroid), othersare located in organs that have distinct, primaryphysiological functions (e.g. heart, gut, kidney).Third, a single endocrine gland may produce morethan one hormone. Fourth, with rare exception,however, a single endocrine cell will synthesize andrelease only one hormone. Fifth, a particular hor-mone may be secreted by more than one endocrinegland. Sixth, a single hormone may elicit several

Chapter 2

Basic Principles and Mechanisms of Endocrinology

MICHAEL R. DESCHENES AND KEIICHIRO DOHI

Page 25: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 9

different physiological responses by binding withseveral different types of target cells. Seventh, anyone hormone, however, elicits only a single res-ponse in any one type of target cell. Eighth, any onetarget cell may respond to numerous hormones,each inducing its own biological response withinthat cell. Ninth, a particular intracellular activity,glycolysis for example, can be regulated by morethan one hormone. Tenth, a target cell’s responsive-ness to a specific hormone may vary according to itsown state of differentiation, presence of other hor-mone, environmental conditions, etc.

While the endocrine system regulates a host ofbiological activities within the target cells of theorganism, the physiological effects of hormones canbe broadly assigned to four areas. These are: (i) thedigestion and metabolism (anabolic and catabolicmechanisms) of food nutrients; (ii) salt and waterbalance; (iii) growth and development; and (iv)reproductive function.

Hormone categorization and synthesis

All of the known hormones can be classified accord-ing to their chemical composition and manner ofsynthesis as either: (i) steroids; (ii) peptides/proteins;or (iii) amines. Steroid hormones are derived fromcholesterol and include the sex steroids (androgens,estrogens, progestins), which are produced in thegonads, as well as the glucocorticoids and mineralo-coids, which are synthesized by the adrenal glands.In humans, the main androgen, or male sex steroid,circulating in the blood is testosterone. Similarly,estrogens are a family of female sex steroids, but in humans estradiol is the primary estrogen, andamong progestins, progesterone dominates. Cortisolis the principal glucocorticoid in humans, andaldosterone is the major mineralocorticoid.

Since the same precursor moleculeacholesterolais used in the formation of all steroid hormones, itis the biosynthetic enzyme pathways present in theendocrine gland that determines the specific steroidthat is mainly produced by that gland. Typically,however, small amounts of a secondary hormoneare concurrently synthesized as some diversionamong the enzyme pathways occurs. For example,the bulk of steroids synthesized by the testes is in

the form of testosterone, yet trace amounts of corti-sol are also manufactured due to a minor presenceof the enzymes that comprise that steroid’s bio-synthetic pathway. In viewing Fig. 2.1, it is easy toappreciate how this diversion among enzymaticpathways can occur.

The rate at which steroid hormones are producedis dictatedaas in all enzymatic pathwaysaby theactivity of the rate limiting enzyme of the pathway,i.e. the enzyme that catalyzes the slowest reactionwithin the cascade. In all steroid hormone pro-duction the rate limiting reaction is the conversionof cholesterol to pregnenolone. Thus, factors thatincrease the rate of steroid production do so primar-ily by accelerating the formation of pregnenolone,and secondarily by amplifying the uptake of cho-lesterol from the blood into the endocrine gland(Rhoades & Pflanzer 2003).

In the endocrine glands that manufacturesteroids, there is no capacity for storage of the newlysynthesized hormone. Accordingly, as the steroid is synthesized, it is secreted into the blood stream,and therefore the rate of the newly formed steroid’srelease into the blood equals the rate of its produc-tion within the endocrine cell.

Peptide and protein hormones are comprised ofchains of amino acids. Should a small number ofamino acids be involved (< 20), the hormone is typ-ically referred to as a peptide hormone, but if 20 ormore amino acids comprise its structure, the chain isconsidered a protein hormone (Goodman 1994).Examples of peptide hormones include oxytocin,vasopressin, and somatostatin. Among the many (~ 100) protein hormones identified to date areinsulin, growth hormone, calcitonin and glucagon.Some of these proteins exist as relatively simple, single chains of amino acids, while others featuredisulfide bonds to connect different regions of thepolypeptide sequence to convey complex tertiarystructure. Some protein hormones are even com-posed of multiple subunits that are bonded togetherto form a single structure.

Regardless of their final structure, all peptide/protein hormones are synthesized within the endo-crine cell in a manner that is consistent with proteinproduction in all cells. That is, peptide/protein hormone precursors are synthesized by ribosomes

Page 26: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

10 chapter 2

ain conjunction with tRNA and mRNAaas muchlonger products than they ultimately appear in theirfinal form. These preprohormones contain signalsequences indicating that the protein is intended forrelease from the cell. Initial modifications of thesemolecules occur in the endoplasmic reticulum,where the ribosomes are attached, and include pro-teolytic events that remove amino acid sequencesaincluding the signal sequencearesulting in shorterchains. The newly formed prohormones then journeyto the Golgi complex where they undergo addi-tional modifications that include further proteolyticcleavage and perhaps the addition of carbohydrate(glycosylation) or phosphate (phosphorylation)groups. Upon completion of these modifications,the Golgi complex pinches off a segment of its membrane to encapsulate the finished hormone in avesicle. This secretory vesicle remains stored in theendocrine cell’s cytoplasm until the cell receives anappropriate signal resulting in an influx of calcium.

The increased intracellular concentration of calciumcauses the membrane of the secretory vesicle to fusewith the cell’s plasma membrane, thus releasing thehormone via exocytosis. Typically the amount ofpeptide/protein hormone stored by the endocrinecell is limited and as a result the signal that stimu-lates the secretion of the cell’s hormone also triggersthe synthesis of additional quantities of that hor-mone (Rhoades & Pflanzer 2003).

The proteolytic cleavage of the prohormone during protein hormone synthesis conveys greatdiversity in the number of hormones produced bythe endocrine system. The same precursor moleculecan undergo differential processing resulting in theassembly of numerous end products. Perhaps thebest example of this is the precursor proopiome-lanocortin (POMC) which contains the amino acidsequences of several peptide/protein hormonesincluding adrenocorticotropic hormone (ACTH), β-endorphin and β-lipotropic hormone, among others

Cholesterol

(side chain cleavage)

Pregnenolone (17α-Hydroxylase)

(17α-Hydroxylase)

(C17–20 Lyase)

(C17–20 Lyase) (Aromatase)

17-Hydroxypregnenolone Dehydroepiandrosterone

AC

AC

DF

AC

DF

Progesterone

(21-Hydroxylase)

11-Deoxycorticosterone

(11β-Hydroxylase)

Corticosterone

(18-Hydroxylase)

18-HydroxysteroidDehydrogenase

Aldosterone

MINERALOCORTICOID

3β-HydroxysteroidDehydrogenase-Isomerase

DF

17-Hydroxyprogesterone

(21-Hydroxylase)

11-Deoxycortisol

(11β-Hydroxylase)

Cortisol

GLUCOCORTICOID

3β-HydroxysteroidDehydrogenase-Isomerase

AC

DF

AC

DF

Androstenedione

(Aromatase)

Estrone

17-HydroxysteroidDehydrogenase

Testosterone

AC

DF17-Hydroxysteroid

Dehydrogenase

Estradiol

(16α-Hydroxylase)

Estriol

3β-HydroxysteroidDehydrogenase-Isomerase

ANDROGEN

ESTROGEN

Fig. 2.1 Biosynthetic pathways of steroid hormones. Involved enzymes are found in parentheses.

Page 27: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 11

(Krieger et al. 1980; Chretian & Seidah 1981). Thespecific cleavage enzymes expressed in the par-ticular endocrine gland synthesizing POMC deter-mines the principal hormone manufactured by thatgland. For example, the anterior pituitary contains a specific set of proteolytic enzymes that will yieldACTH as its primary end product from the POMCprohormone. In contrast, neurons within the brainthat produce POMC possess enzymes that cleavethe molecule in such a way that β-endorphin is mainlysecreted. This differential processing of POMC alsooccurs in the placenta, reproductive organs, gas-trointestinal tract and lung (Liotta et al. 1982;Margioris et al. 1982). This gland specific expressionof different hormones from a common precursormoleculeabased upon the enzyme profile presentais not unlike the biosynthesis of steroids.

Amine hormonesaalso referred to as amino acidderivativesaare those that use the amino acid tyrosine as their initial precursor. Included in thiscategory are the thyroid hormones (thyroxine [T4]and triiodothyronine [T3]) and the catecholamines(epinephrine and norepinephrine). Despite sharingthe same precursor molecule, the thyroid hormonesand catecholamines differ in many respects includ-ing their synthesis, transport through the bloodstream and mechanism of action at target cells. Herethe amine hormones will be addressed separately indescribing their synthesis.

Thyroid hormone formation is dependent uponthe uptake of both tyrosine and the mineral iodidefrom the blood into the follicular cells of the thyroidgland. Tyrosine is used as a backbone for the manu-facture of thyroglobulin which is a large glyco-protein that is stored in large amounts within thefollicular cells. With the uptake of iodide from the blood, the tyrosine residues of thyroglobulinbecome iodinated through a multistep reaction, ultimately forming either T4 or T3, depending on thenumber of iodide ions that bind with thyroglobulin.Initially, thyroglobulin reacts with either one or twoiodide ions resulting in the production of eithermonoiodotyrosine (MIT), or diiodotyrosine (DIT),respectively. In the next step of the enzymatic path-way of thyroid hormone synthesis, two iodideatoms are added to the thyroglobulin molecule converting MIT to T3 and DIT to T4. At this point the

thyroid hormones are part of the larger thyroglobu-lin structure that is stored within the glandular cell.Upon stimulation to release thyroid hormone, pro-teolytic enzymes within the follicular cell breakdown the stored thyroglobulin liberating the T3 andT4 hormones intact, which are then released into thecirculation.

The catecholaminesaepinephrine and norepine-phrineaalso use tyrosine as their initial precursor,but are produced in the medulla region of theadrenal glands. This adrenomedullary tissue isactually a modified component of the sympathetic,or excitatory, branch of the autocrine nervous sys-tem. In fact, the adrenal medulla directly receivesinput from nerve terminals of the sympathetic nerv-ous system serving as an example of neuroendo-crine function.

Catecholamine synthesis occurs in the chroma-ffin cells of the adrenal medulla via a multistep biosynthetic pathway. First, tyrosine is converted to 3,4-dihydroxyphenylalanine (dopa) by tyrosinehydroxylase which acts as the rate limiting step inthe production of catecholamines. Dopa is then converted to dopamine, which is then transformedinto norepinephrine, most of which is methylatedby the enzyme phenylethanolamine–N–methyltransferase (PNMT) resulting in the formation ofepinephrine. Both norepinephrine and epinephrineare considered catecholamines, but the stoichiome-try of their synthesis and release from the adrenalgland is 1 : 4. Although circulating levels of nore-pinephrine exceed those of epinephrine, the bulk of this norepinephrine originates from the sym-pathetic nervous system where it functions as a neurotransmitter and ‘spills over’ into the blood.Epinephrine, however, is the primary catecholam-ine hormone circulating in the bloodstream (Hedgeet al. 1987).

Upon their production, the same glandular cellsthat synthesize the catecholamines store them aschromaffin granules. Stimulation by the sympatheticnervous system results both in the secretion of cate-cholamines through typical exocytotic actions, andincreased activation of tyrosine hydroxylase affect-ing a greater rate of catecholamine synthesis by thechromaffin cells, so that intracellular catecholaminedepots can be replaced (Rhoades & Pflanzer 2003).

Page 28: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

12 chapter 2

Regulation of hormone secretion

The effect of a hormone on its target tissue is propor-tional to its concentration within the bloodstream.Several factors combine to determine the level ofany biologically active hormone in the circulation.These variables include: (i) rate of secretion from the endocrine gland into the circulatory system; (ii)for some hormones, i.e. thyroid hormones, rate ofactivationathe conversion of T4 to T3awithin theblood; (iii) for lipophilic hormones, i.e. steroid andthyroid hormones, degree of binding to plasma pro-teins; and (iv) rate of inactivation and clearancefrom the blood. Of these factors it is the first-rate ofrelease into the bloodathat is the principal deter-minant of circulating hormone levels, particularlyunder non-exercising conditions (Sherwood 2004).

In general, there are two modes of hormonalrelease into the bloodstream (Kelly 1985). Constitut-ive release refers to a continual, moderate dischargeof endocrine agent into the circulation. In this releasemechanism, hormone exits as it is synthesized; thereis no storage capacity within the endocrine gland.Consequently, upon receipt of stimulatory signal,synthetic pathways increase their activity andnewly formed hormone is directly released into thecirculation via passive diffusion through the cell’splasma membrane. This release pattern governs circulating levels of steroid and thyroid hormones,which like the cell’s plasma membrane, are lipo-philic in nature. Constitutive secretion is mediatedby alterations in the phosphorylation status of pro-teins acting as enzymes in endocrine biosyntheticpathways.

Regulated release is the second mode of delivery ofhormone from the endocrine gland to the blood-stream. In this case, the rates of protein synthesisand release are not directly coupled, as they are inconstitutive release. Rather, endocrine glands thatemploy regulated release possess the ability to storenewly synthesized hormone. It should be noted,however, that even in this type of endocrine glandstorage potential is limited. Indeed, for any particu-lar hormone, it is rare for even a day’s supply to bestored and available for release (Baulieu 1990).

In regulated release, a stimulus causes the exo-cytotic release of prepackaged vesicles containing

hormone. In most cases, to assure a ready supply ofhormone, the same stimulus that evokes the releaseof stored hormone also activates enzymatic path-ways that synthesize that hormone. An influx of calcium into the endocrine cell’s cytosol usually pre-cedes the release of stored hormone and activationof biosynthetic pathways. This regulated form ofrelease is evident in the secretion of peptide/proteinhormones, as well as catecholamines.

With both constitutive and regulated hormonalrelease, the stimuli that govern secretion are typic-ally: (i) changes in plasma nutrient or ion concentra-tions; (ii) neurotransmitters released from neuronsonto endocrine cells; or (iii) the binding of hormonesreleased by other endocrine glands. Generally, thesestimuli do not operate independently of each other.To the contrary, alterations in endocrine secretionusually are a function of receiving input from morethan one type of stimulus.

The responsiveness of endocrine glands to thesestimuli is contingent upon a sensitive and effectivefeedback system that conveys information from target tissues back to the hormone releasing organ.The most prevalent form of communication regulat-ing secretory rates of endocrine glands is negativefeedback. This type of feedback occurs when theactivity of one system (endocrine gland) adjusts tonegate, or offset, a change in another system (targettissue), thereby re-establishing homeostasis. Forexample, an elevation in blood glucose triggers thesecretion of insulin by the pancreas. The increasedblood-borne insulin level expedites the uptake ofglucose into fat and muscle cellsatarget cells ofinsulinaresulting in a normalization of glucose con-centration in the blood.

There are even multiple forms of negative feed-back regulation controlling hormone secretion. Therelease of several important hormones is directed by the hypothalamic–pituitary axis. A small regionat the base of the brain, the hypothalamus producesseveral releasing hormones that are transported bya portal blood supply to the anterior segment of thepituitary gland. In this example of neuroendocrinefunction, the hypothalamus delivers releasing hor-mones to the pituitary which, in turn, alters its rateof secretion of a number of hormones into the sys-temic circulation where they can be transported

Page 29: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 13

throughout the organism. These pituitary hormonesmay directly bring about effects on target tissues, orthey may react with a third gland in the axis, therebyinfluencing this gland’s secretion of hormone intothe bloodstream.

Regarding negative feedback, this neuroendo-crine axis can display both ‘short-loop’ and ‘long-loop’ regulation (Vander et al. 2001). In an exampleof short-loop negative feedback, high levels of pro-lactin in the bloodstream are detected by thehypothalamus causing it to increase its own secre-tion of dopamine to the pituitary, thus curtailingthat gland’s release of prolactin into the circula-tion. In the three organ neuroendocrine axis, long-loop negative feedback is also evident. To illustratethis feedback mechanism, the regulation of cortisolsecretion will be discussed. The axis involved features the hypothalamus, pituitary, and adrenalcortex. The hypothalamus delivers corticotropin-releasing hormone (CRH) into the portal blood sys-tem where it is carried to the anterior pituitary.Upon binding of CRH, the pituitary releases ACTHinto the systemic bloodstream so that it can be deliv-ered to the adrenal glands. In the cortex region of theadrenalarecall that the adrenal medulla producescatecholaminesathe binding of ACTH triggers therelease of cortisol into the systemic circulation sothat it can elicit glucocorticoid effects on target tissues such as the liver, skeletal muscle and adip-ose. In this case, negative feedback occurs when theelevation in blood-borne cortisol blunts the releaseof ACTH from the pituitary and/or the secretion of CRH from the hypothalamus. As an example ofthe exquisite integration of signals delivered andreceived throughout the endocrine system, the hormone secretion of a multiorgan endocrine axiscan be regulated both by short-loop and long-loopfeedback (Vander et al. 2001). Illustrations of short-and long-loop negative feedback are displayed in Fig. 2.2.

Although negative feedback is much more preval-ent, positive feedback is also used to adjust hormonesecretion. In positive feedback, the hormone inducedchange in the biological activity of the target tissueis monitored by the endocrine gland that initiallyreleased the hormone. Should the target tissue’sresponse be insufficient in magnitude, the endo-

crine gland secretes additional hormone until the biological process it governs is adequate. Anexample of positive feedback is the regulation ofendocrine function during childbirth. Oxytocinreleased by the posterior aspect of the pituitarygland stimulates muscle contraction of the uterus.As the birthing process continues and stronger uterine contractions are required, the activity of theuterus signals the pituitary to increase its secretionof oxytocin, thus amplifying the strength and fre-quency of uterine contractions enabling the com-pletion of parturition.

Although the primary function of the endocrinesystem is to maintain homeostasis, changes in thebody’s internal and external environments are notthe only regulators of hormone secretion. Indeed,circulating concentrations of most hormones dis-play predictable fluctuations, or rhythms, over agiven period of time. The best studied endocrinerhythm is the circadian, or diurnal, rhythm. ‘Circadian’refers to the pattern of peaks and troughs through-out the roughly 24-h solar day, while ‘diurnal’alludes to the day/night oscillations of hormone

Stimulus

Portal vasculature

Second gland(anterior pituitary)

releases hormone into . . .

Systemic circulation

Long-loopfeedback

Short-loopfeedback

Third gland(adrenals, gonads, etc.)

releases hormone into . . .

Systemic circulation

First gland(hypothalamus)

releases hormone into . . .

Fig. 2.2 Illustration of short-loop and long-loop negativefeedback in the regulation of hormone release.

Page 30: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

14 chapter 2

secretion. Often these two terms are used inter-changeably. These natural, preprogrammed rhythmsof endocrine release originate in the suprachias-matic nucleus region within the hypothalamus. This pacemaker regulates the secretion of hormonesbased upon its own internal clock, and elicits releasepatterns that are specific to each hormone. Forexample, cortisol levels in the blood are highest inthe morning, while growth hormone peaks duringthe night-time hours (Illnerova et al. 2000).

In addition to variation over the 24-h day, hor-mone secretion often displays a regular pulsatilepattern sometimes referred to as an ‘ultradian’rhythm that is superimposed on the underlying cir-cadian rhythm. These episodic bursts of hormonerelease also appear to be directed by the hypotha-lamus and may have important physiological con-sequences. For example, it has been demonstratedthat even when total dosage is the same, glucoseuptake into target tissue is enhanced when insulin is delivered in pulses compared to a steady, non-pulsatile pattern (Porksen 2002).

Although not as well studied in humans as in animals, hormone secretion is also known to varyseasonally. These ‘circannual’ rhythms correspondto changes in the number of daylight hours, whichare sensed by the pineal gland within the centralnervous system (Short 1985). This gland, oftenalluded to as the ‘third eye’ due to its photosensit-ivity, adjusts the amount of melatonin it secretes inresponse to alterations in total daylight hours. Inanimals demonstrating seasonal breeding behavior,the production of gonadotropins is modulated bythese circannual variations in melatonin production(Tamarkin et al. 1985). Other seasonal dependentbehaviors, such as hibernation, migration and evenchanges in the color of fur, are guided by the pre-dictable oscillations in circulating melatonin levels.In humans, increased melatonin production, whichoccurs as the number of daylight hours decreases,has been associated with altered mood states andeven depression (Lewy et al. 1987). More recently ithas been established that in all mammalsaincludinghumansamelatonin plays a major role in ensuringproper circadian rhythmicity by interacting withand influencing the suprachiasmatic nucleus (Pevetet al. 2002).

Hormone transport in the blood

Each hormone is released from the endocrine glandthat produced it into the venous capillaries sur-rounding that gland. After passing through thelungs and heart, the hormone enters the general sys-temic circulation so that it may travel throughoutthe body. While traveling in the blood, a very smallamount of hormone may be found adsorbed ontothe plasma membrane of red blood cells, but mostlythey are dissolved in the plasma (peptide/proteinhormones), or bound to plasma proteins (steroids,thyroid hormones). Due to their chemical structurethe peptide/protein hormones are hydrophilic innature and readily dissolve in the blood’s plasma.But once in the bloodstream these hormones areexposed to numerous proteolytic enzymes thatbreak them down, thus preventing them from inter-acting with their target tissues. Recall, however, thatendocrine glands that synthesize peptide/proteinhormones are able to store them and secrete themupon receiving a stimulatory signal indicating aneed for that particular hormone.

In contrast to the peptide/protein hormones,steroid hormones are hydrophobic in structure, andas a result, are not soluble in the plasma. Con-sequently, the vast majority of these hormones (> 95%) travel through the bloodstream bound tovarious plasma proteins. The amine hormones alsoare bound to proteins as they travel through theblood, albeit to various degrees. Only about 50% ofblood-borne catecholamines are bound to carriers,while a much higher proportion (~ 99%) of thyroidhormones, which are hydrophobic in structure, arein the bound state.

Steroids may either bind to a plasma protein dis-playing high specificity for a particular hormone(Table 2.1), or with lower specificity and affinity to albumin and transthyretin, which are abundantcirculating proteins. All carrier proteins, the highspecificity transport proteins and the ubiquitousalbumin and transthyretin, are synthesized by theliver before being released into the blood (Rhoades& Plfanzer 2003). The low affinity albumin andtransthyretin molecules contain several bindingsites which can be bound not only by hormones butalso by other low molecular weight substances

Page 31: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 15

found in the blood. In contrast, the high specificitytransport proteins typically possess a single bindingsite which is configured to interact with a specifichormone, and displays greater affinity at its ligandbinding site than does albumin or transthyretin(Baulieu 1990). The percentage of circulating hor-mone bound to albumin versus a specific transportprotein differs among hydrophobic hormones. Forexample, equal amounts of bound aldosterone arecomplexed with specific binding proteins and albu-min or transthyretin. But only 10% of circulatingcortisol is bound to albumin or transthyretin, whilethe vast majority (> 85%) is bound to corticosteroidbinding globulin.

Whether bound to albumin, transthyretin, or aspecific transport protein, only the small amount ofsteroid or thyroid hormone that is unbound or ‘free’is capable of binding with receptors sites expressedby target tissue. Thus, only the free portion of thesehormones is able to stimulate biological activitywithin target cells. As this free steroid or thyroidhormone interacts with target tissue, some of thebound fraction dissociates from binding proteins,thereby forming a dynamic equilibrium betweenthe bound and unbound fractions that is influencedby the need to maintain the organism’s homeostasis.In effect, the bound fraction serves as a buffer tosupply hormone to the target tissue as needed. Thisis presumed to compensate for the fact that unlikepeptide/protein hormones, the glands that synthes-ize steroids and thyroid hormones are unable tostore newly formed hormone to be released upondemand. Indeed, it is no longer considered that theprimary function of transport proteins is to over-

come the problem of insolubility in the plasma. Ithas been demonstrated that due to their small size,most steroid and thyroid hormones readily dissolvein the blood despite their hydrophobic chemicalstructure (Kronenberg et al. 2003). This suggests thatthe binding proteins mainly act to provide a buffer-ing or storage capacity, and to prevent the small,intact hormones from passing from the blood intothe renal tubules of the kidneys where they can beprematurely excreted from the body. In the case ofinsulin-like growth factors, which are hydrophilicin structure, specific binding proteins act to directlymediate the biological effect imparted by the hor-mone/receptor complex at the target cell (Firth &Baxter 2002).

Metabolic clearance of hormones

After entering the circulation most hormones are rapidly degraded and feature half-lives of nomore than 30 min, although this can vary greatly.Catecholamines, for instance, have half-lives of onlyseconds, while thyroid hormones exhibit half-livesof several days. The length of any hormone’s half-life in the bloodstream is dependent upon themetabolism and/or clearance from the circulation.As with transport, the chemical composition of thehormone will determine the specific method of itsremoval from the circulation, but in general, all hormones are primarily broken down within theliver and eliminated via the renal system. Peptide/protein hormones and catecholamines are typic-ally degraded by proteolytic enzymes in the bloodand the resultant amino acids are excreted through

Table 2.1 Examples of high specificity and affinity blood-borne hormone binding proteins.

Name Abbreviation/alternate name Hormone(s) bound

Sex hormone binding globulin SHBG/sex steroid binding protein Testosterone, estradiolCorticosteroid binding globulin CBG/transcortin Glucocorticoids, progesterone,

aldosterone (minor amounts)Thyroxine binding globulin TBG Triiodothyronine (T3),

tetraiodothyronine (T4)Growth hormone binding protein GHBP Growth hormoneInsulin-like growth factor IGFBPs IGF-I, IGF-II

binding proteins

Page 32: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

16 chapter 2

the urinary tract. In contrast, steroid and thyroid hormones are resilient to circulating proteolyticenzymes, and they are mainly degraded in the liverthrough a series of reducing reactions which increasethe solubility of these hydrophobic molecules. Since steroids are small in structure, the improvedsolubility then allows the majority of them to beeliminated via the urinary tract, although a smallportion is secreted directly from the liver into thebile (Rhoades & Pflanzer 2003). Another, but lessprevalent, mode of hormonal clearance is termed‘receptor-mediated endocytosis’. In this processsome larger protein hormones such as insulin areinternalized by the target cells after they have boundwith receptors and elicited a response. After beingengulfed by the target cell, the hormone is separ-ated from its receptor and degraded by proteolyticenzymes located within the cell’s cytoplasm.

Mechanism of action at target cells

General principles

The biological action(s) elicited by a hormone at itstarget cell is initiated by the binding of the hormoneto its receptor produced by the cell. Indeed, thisreaction between hormone and receptor lies at thefoundation of the endocrine system. In this manneran endocrine gland may release a hormone into thegeneral circulation where it will perfuse all tissueswithin the body, but will exert its influence only on those cells that express receptors specific for thathormone. Typically, a target cell will express 2000–100 000 receptors for any one hormone (Guyton &Hall 1996).

A receptor is a proteinasometimes a glycopro-teinathat possesses one or more binding sites forligand (hormone) and an effector, or active, site thattriggers a biological response in the cell expressingthat receptor. Each type of receptor displays aunique three-dimensional structure that is compli-mentary to the structure of a specific hormone, thusconferring the property of specificity to the receptor.The complimentary chemical structures apparent inthe hormone and its receptor allow them to recog-nize each other and interact in what is sometimesreferred to as a ‘lock and key’ formation. It is pos-

sible, however, that two hormones with very similarchemical structures, for example insulin and insulin-like growth factor, will cross-react with each other’sreceptors. Under normal conditions and hormoneconcentrations, such cross-reactivity is minimal andthe resultant cellular activity is negligible.

The strength of the bond formed between the hor-mone and its receptor is described as the affinityof the ligand for its binding site. The affinity of thehormone–receptor complex determines how easilythe bond can be broken. In high affinity reactions,much disturbance to environmental conditions, for example pH, temperature, etc., is necessary todisrupt the hormone–receptor complex. In contrast,low affinity bonds are easily broken and oftenrequire no change at all in the surrounding milieu.The cross-reactivity described above displays thislow affinity binding, which is why those hormone–receptor complexes typically result in very little biological response in the cells where they occur.

A dynamic equilibrium exists between unboundhormone in the blood perfusing the target tissueand the hormone bound to the tissue’s receptors.This equilibrium is essential to the endocrine sys-tem’s regulatory function. Consistent with the lawsof mass action, the greater the amount of hormoneavailable to the targetacirculating concentrationa

the greater the probability that receptor sites will beoccupied by hormone, and thus the greater the bio-logical response evident at the target. More directlystated, the target cell’s change in biological activityis proportional to the number of hormone–receptorcomplexes formed, which is dictated by the hor-mone’s concentration in the bloodstream. Saturationrefers to the fraction of the target cell’s receptors thatis bound to hormone, and in physiological terms,this is critical in determining the response of the cellto the hormone. Two factors primarily account forthe degree of saturation of a cell’s binding sites: theconcentration of unbound circulating hormone andthe affinity of receptors for that hormone.

In considering the importance of saturation inregulating a target cell’s response to a hormone, it isworth mentioning that, even at maximal biologicalresponse, not all of the cell’s receptors for that hormone are bound. This observation has lead tothe ‘spare receptors’ concept of endocrine function.

Page 33: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 17

That is, 100% occupancy of receptors is not neces-sary to elicit maximal biological response, suggest-ing that the cell expresses more receptors thannecessary. This ‘overexpression’ of receptors is, infact, appropriate when one recalls that the forma-tion of hormone–receptor complexes is governed by the rules of statistical probability. There are noforces of attraction that bring complimentary hor-mones and receptors together, although affinity sustains the bond once formed. There is simply achance that circulating hormone and matchingbinding site will come into contact with each other,and then form a complex. Thus to enhance the prob-ability that such random contacts will occur, targetcells express more receptors than are needed tostimulate even maximal response. The expression of these ‘spare receptors’ is to be considered espe-cially efficient and sensible given the very low con-centrations at which hormones travel in the blood(10–8–10–12 mol·L–1).

Competition describes the process whereby dif-ferent ligandsadisplaying similar chemical structureamay compete for binding at the same receptor.Under normal physiological conditions, little com-petition exists between hormones traveling throughthe bloodstream. The potential for such competi-tion, however, enables the pharmacological man-agement of some endocrine disorders, as drugsacting as antagonists may compete with endogen-ous hormones that are over-secreted for bindingsites, thereby preventing an inappropriately ampli-fied endocrine response. Also, the measurement of serum or plasma concentrations of hormones ispredicated upon competition as endogenously pro-duced hormones and labeled antigens vie for agiven number of binding sitesaantibodiesapro-vided in the assay.

Although, in general, the concentration of anyhormone present in the bloodstream dictates thedegree of the biological response elicited by thathormone, the target cell is capable of fine tuning itsresponse by adjusting the numberaand perhapsaffinityaof the receptors it expresses for that hor-mone. That is to ensure its proper function, targettissue may ‘up-regulate’ or ‘down-regulate’ its hor-monal receptors to maintain its delicate homeostaticbalance. The advantages of controlling the number

of available binding sites is obvious when one con-siders that several types of cells, each with its ownneeds and homeostatic challenges, are sensitive tothe same hormone that is secreted into the systemiccirculation. For example, a disturbance in the envir-onment of one cell type may trigger increasedrelease of a particular hormone to counter it. Butanother cell that is also responsive to that hormonemay be at risk of homeostatic imbalance as a resultof the greater levels of that hormone. To preventthis, the cell may down-regulate, or temporarilydecrease the number of receptors it makes availableto bind the hormone. This would decrease the prob-ability of hormone–receptor complex formation and dampen the cell’s sensitivity to the hormone,thus maintaining its own homeostasis. Such adjust-ment in receptor number and/or affinity allowseach type of target tissue to respond in a mannerappropriate to its own needs when exposed to agiven amount of hormone traveling throughout the entire organism. More long-term up- or down-regulation of receptor expression is evident withdramatic alterations in the production of hormonethrough surgical removal of endocrine glands(Dahlberg et al. 1981), disease (Potier et al. 2002;Pedersen & Vedickis 2003), or even as an adaptationto chronic exercise training (Tchaikovsky et al. 1986;Deschenes et al. 1994).

Not only do target cells express receptors for ahost of different hormones so that various cellularfunctions can be regulated by the endocrine sys-tem, the same cell may respond to more than onehormone which affects the same biological pro-cess within the cell. This phenomenon is known asredundancy, but should not be considered a super-fluous or inefficient means of controlling cellularphysiology. For example, hepatocytes are sensit-ive to various hormonesaincluding glucagon andepinephrineathat stimulate glycogenolysis and therelease of glucose into the blood. However, differentconditions may account for the need for additionalcirculating glucose. Missing a meal may result indepressed glucose levels in the bloodstream, whichwould be detected by the pancreas which wouldrespond by secreting glucagon to break down glyco-gen stores in the liver. Epinephrine, too, stimulatesglycogenolysis and the release of glucose from the

Page 34: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

18 chapter 2

liver into the circulation. However, this is part of an overall ‘fight or flight’ response elicited by thesympathetic nervous system and the adrenal secre-tion of epinephrine that includes increased heartrate, blood pressure and sweat rate. Obviously,these responses would be inappropriate to addressa simple fasting induced state of hypoglycemia.Seen in this context, redundancy in the expression ofreceptors for the glycogenolytic hormones does notappear wasteful, but rather a sophisticated approachto managing cellular homeostasis in the face ofmany different challenges.

Binding of hormone to receptor

The biological response attributed to a hormonemust be preceded by the binding of that hormonewith its receptors located at its target cells. In thisscenario, it is best to view the receptor as a mediatorthat transducts the extracellular message carried bythe hormone to an intracellular signal that ulti-mately leads to a specific cellular response.

At each hormonal receptor characterized to date,only a single binding site exists so that at any time,only a single molecule of hormone can occupy abinding site to form a hormone–receptor complex.Unlike enzymatic reactions where the substrate isaltered by binding with enzyme, the hormone isunaffected by the receptor to which it is bound. It is also noteworthy that the hormone–receptor com-plex is formed by a non-covalent bond that isreversible, and thus transient.

Upon binding with its hormone, the receptorundergoes a modification to its three-dimensionalstructure. It is this conformational shift that activ-ates the effector site of the receptor, setting forth a cascade of events which result in the cellularresponse that is ascribed to the involved hormone. It is important to recognize that while the binding of hormone to receptor is reversible, the biologicalevents initiated by the formation of hormone–receptor complex continue for some time after thedissolution of that complex. On the other hand, thishormone induced cellular response is limited in the absence of additional hormone–receptor com-plex formation. Consequently, if the target cell is toremain activated for an extended period of time in

its attempt to achieve homeostasis, the availabilityof hormone molecules must be sustained so thatnew molecules may bind with freshly unoccupiedreceptors.

Types of receptors and post-receptor actions

At all target cells and with all hormones, the bio-logical response evoked by a hormone must be preceded by the binding of the ligand (hormonemolecule) with its specific receptor expressed by the hormone sensitive cell. Despite the plethora ofhormones produced by the endocrine system andthe many cell types that respond to these hormones,all receptors can be broadly categorized as eithermembrane bound or intracellular receptors. Mem-brane bound receptors are constituents of the tar-get cell’s plasma membrane and bind to peptide/protein hormones, as well as catecholamines. Asindicated by their name, intracellular receptors are located within the cell, and bind to steroid and thyroid hormones which are small lipophilicmolecules that easily diffuse across the plasmamembrane to enter the cell.

intracellular receptors

In their unbound state there is some disparity in the precise localization of intracellular receptors.Initially, it was believed that unbound intracellularreceptors were found in the cell’s cytosol and that,upon hormone binding, the hormone–receptorcomplex was translocated into the nucleus. How-ever, it is now understood that even in the unboundstate, intracellular receptors for most steroid hor-mones are located within the nucleus. An exceptionis the glucocorticoid receptor, which when unoccu-pied is anchored to the cytoplasmic exterior of thenucleus (Lazar 2003). One feature that receptors oflipophilic hormones have in common is that theyare considered inactive when they are not boundwith ligand. Characteristic of this inactive state isthat these receptors are found in association withheat shock proteins (HSPs), particularly HSP90 (Joabet al. 1984; Catelli et al. 1985). Hormone binding tothe receptor results in a dissociation of the receptorfrom these chaperone proteins and dimerization of

Page 35: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 19

newly liberated receptors. Upon this ‘activation’,both cytoplasmic and nuclear receptors are trans-located to nuclear DNA, in order to interact withhormone response elements (HREs). Once adheredto these HREs, the hormone–receptor complex regulates the transcription of specific genes on theDNA. Accordingly, intracellular receptors are con-sidered members of the larger family of transcrip-tion regulatory factors.

Determining the amino acid sequence of theseintracellular receptors has provided much insightinto the mechanisms of their interactions with hor-mones and DNA. At the C-terminal segment of the protein structure is a sequence of about 250amino acids that are lipophilic in nature and form astructural pocket capable of binding steroid andthyroid hormones. The unique sequence of aminoacids comprising this pocket determines the recep-tor’s specificity for a particular hormone.

Binding of the hormone–receptor complex tospecific HREs linked to hormone sensitive genes isdirected by a DNA binding domain consisting ofapproximately 70 amino acids that are also locatedat the C-terminus of the receptor protein. Commonto all DNA binding domains are two highly con-served regions referred to as ‘zinc fingers’ whichdirectly interact with DNA (Scheidereit et al. 1986).The ensuing genetic transcription is carried out by a transcription complex that, in addition to the hormone–receptor complex, also includes ‘generaltranscription factors’ and ‘positively acting cofactors’(McKenna et al. 1999). These coactivators enhancethe rate of transcription, at least in part, by unwind-ing the DNA double helix and stimulating RNApolymerase activity (Kuo & Allis 1998).

Thus the cascade of events that result in theactions of steroid or thyroid hormones at target cellsincludes:1 entrance of hormone via simple diffusion;2 formation of hormone–receptor complex;3 dissociation from HSPs and receptor dimerization;4 translocation to DNA;5 binding to HREs;6 assembly of transcription complexes;7 synthesis of gene specific mRNA;8 within cytosol, translation of proteins coded bymRNA.

As an illustration of the efficiency and integration ofthe endocrine system, it appears that the actions of asingle lipohilic hormone bound to its intracellularreceptors can regulate the transcriptional activity of several genes, altering the synthesis of severalproteins. Yet this occurs in a co-ordinated manner in that each of the newly synthesized proteins par-ticipates in a single, overall biological responsewithin the cell. For example, aldosterone functionsto maintain proper sodium levels within the body. Itdoes this by stimulating the reabsorption of sodiumfrom the renal tubules when blood concentrations of that mineral are low. To achieve this end, aldos-terone increases the synthesis not only of sodiumchannels and sodium pumps within the membranesof the tubules, but also the enzymes that manufac-ture the adenosine triphosphate (ATP) used bythose pumps. This synchronized production of sev-eral proteins subserves the reabsorption of sodiumfrom the renal filtrate in order to maintain properosmolality and water balance within the body.

Since the mechanism(s) of action of intracellularendocrine receptors involves the management ofprotein production, the biological activities stimu-lated by steroid and thyroid hormones are typicallyslow to occur and are long lasting. Recent evidence,however, suggests that in some cases lipophilic hormones induce fast acting responses of shortduration in target cells, without any change in pro-tein expression (Oichinik et al. 1991). Although theexact machinations of these rapid responses remainundefined, it appears that they are triggered by theinteraction of steroid hormone with receptorslocated on the plasma membrane of the target cell,rather than with intracellular binding sites.

membrane bound receptors

As opposed to receptors that bind with lipophilichormones which easily diffuse across the target cell’s membrane to enter the cell, receptors for lipo-phobic hormones (peptides/proteins) are locatedon the target’s plasmalemma since these hormonesare not able to enter the cell. And because forma-tion of the hormone–receptor complex occurs at the membrane’s extracellular surface, the resultantintracellular activity is triggered by a pre-existing

Page 36: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

20 chapter 2

signal transduction mechanism that provides arapid response. In short, because membrane boundreceptors are linked to second messenger (the hor-mone acts as the first message) systems that arealready in placeasimply needing to be activated byformation of hormone–receptor complexathe cellu-lar response elicited is quickly turned ‘on’ and canbe just as quickly turned ‘off’.

It is the chemical structure of membrane boundreceptors that enables the forwarding of an extracel-lular message presented by a hormone molecule to pre-existing signal transduction pathways withinthe cell. All membrane receptors are comprised ofthree distinct sections, even though the receptoritself is generally a single polypeptide chain (Spiegelet al. 2003). The extracellular region is located at the N-terminal end of the chain, and consistentlyexpresses glycosylation sites. The carbohydrateresidues found at those sites appear to be involvedin hormone binding, which occurs specifically incysteine rich pockets. Each of the receptor’s trans-membrane segmentsamembrane receptors typicallyfeature several membrane spanning regionsa

consists of about 25 amino acids that are lipophilicin nature and form a helical structure. The intracellu-lar region is at the C-terminal end of the chain and isresponsible for the effector function of the receptor.Generally, the intracellular regionawhich is com-posed of lipophobic amino acidsacontains regulat-ory elements including phosphorylation sites.

Although the actions of membrane bound recep-tors are carried out by numerous second messengerpathways, these post-receptor mechanisms can becategorized as: (i) ligand gated channels; (ii) recep-tor bound kinases; (iii) receptor bound guanylatecyclase; (iv) cytokine receptors; or (v) G protein coupled receptors. In the first three categories, whenhormone binding occurs, the receptor itself directlystimulates cellular responses, but in cytokine recep-tors and G protein coupled receptors, the recep-tor activates another molecule within the target cellto carry out the final response attributed to theinvolved hormone.

In the case of ligand gated channels, the receptorexpresses not only an extracellular hormone bind-ing site, but also a channel within the membranespanning regions that allows specific ions to trans-

port when opened. The opening of the channeloccurs upon binding of ligand, resulting in a con-formational shift within the membrane spanninghelices enabling specific ions to cross the plasmal-emma to enter, or exit, the cell. This type of receptoris found on excitable cells, i.e. neurons and myocytes,and the movement of ions electrically stimulates aresponse in the target cell.

The best studied and most common receptor that is directly linked with kinase activity involvestyrosine kinase. Within the human genome, approx-imately 100 receptor tyrosine kinases have beenidentified (Spiegel et al. 2003). Although character-istics of the membrane spanning region are commonto all of them, a large degree of variability exists atthe extracellular region accounting for the diversityof ligands that interact with these receptors. Unlikemost membrane bound receptors, members of thetyrosine kinase family of receptors span the mem-brane but once. Yet similar to many other ligandreceptors, these proteins dimerize upon binding ofhormone, a key step in activating the receptors.Upon this activation, a specific region of the recep-tor’s intracellular domain phosphorylates tyrosineresidues of enzymes within the cell’s interior, thuseliciting the desired response of the cell. Moreover,the activated receptor is capable of phosphorylatingits own tyrosine residues located on its cytoplasmicregion. This ‘autophosphorylation’ allows the ac-tivation state of the receptor to persist, thereforeamplifying the signal carried by the hormone. Acombination of the dissolution of the hormone–receptor complex at the surface of the cell’s exteriorand the activity of intracellular phosphatases, whichde-phosphorylate enzymes, terminate the hormoneinduced biological response within the cell.

Other receptor bound kinases, less prevalent than tyrosine kinase receptors, demonstrate similarmechanisms of activity in that they phosphorylatepre-existing enzymes found within the target’scytoplasm to alter cellular function. Both serine and threonine residues of enzymes residing withinthe cell can be phosphorylated by membrane receptors occupied by hormone. Again, cellular res-ponses cease when hormone–receptor complexesare no longer formed, and when phosphatase activity de-phosphorylates activated enzymes. This

Page 37: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 21

demonstrates a common theme to the mechanism of membrane receptor activity; cellular response isturned ‘off’ both at the extracellular and intracellu-lar levels.

Receptors that mediate their responses via guany-late cyclase compose the final category of membranereceptors where a component of the receptor pro-tein itself stimulates cellular response. This categoryis less ubiquitous than the other types of membranereceptors; it appears that only atrial natriuretic factorutilizes this type of receptor to regulate the activityof its target cells. Here, again, the transduction of thefirst messenger (hormone) to intracellular activity is restricted to the membrane molecule. After bind-ing of ligand to the extracellular domain, a con-formational shift occurs throughout the structureresulting in the activation of guanylate cyclase; an enzyme that is a constituent of the receptor’sintracellular region. Upon activation, this enzymeconverts guanosine triphosphate (GTP) into cyclicguanosine monophosphate (cGMP). The newlyformed cGMP is linked to a nucleotide dependentprotein kinase which, in turn, phosphorylatesenzymes within the cell’s cytoplasm.

Cytokine receptors are quite similar to tyrosinekinase receptors in their mechanism of action: thesereceptors also employ tyrosine kinases to evoke cellular responses. However, a major differenceexists in that cytokine receptors do not express thekinase enzyme itself in their intracellular tails.Rather, the enzyme is structurally uncoupled fromthe cytokine receptor molecule. Members of thecytokine receptor familyawhich include growthhormone and prolactin receptorsaare comprised ofmultiple subunits. Upon binding of ligand, thesereceptors form oligomers and activate the janus tyro-sine kinases, or JAKs, that are located in close prox-imity to the receptor along the cytoplasmic lining of the cell’s plasmalemma (Heim 1999). Cytokinereceptors themselves display no enzymatic activity(Argetsinger et al. 1993). It appears that the activa-tion of JAKs occurs as a result of the drawingtogether of neighboring JAKs when oligomers ofhormone–receptor complexes form, allowing theJAKs to transphosphorylate each other’s tyrosineresidues. The activated kinases then phosphorylatecytosolic enzymes which, ultimately, carry out the

hormone’s message. As previously described, hor-mone induced cellular responses are halted by the unbinding of hormone from receptor, as well asthe activity of intracellular phosphatases whichinactivate enzymes within the target.

The most prevalent category of membrane boundreceptors is the family of G protein coupled receptors.Over a thousand different ligands execute their biological activities via these receptors (Spiegel et al.2003). In all cases, however, the G protein and itscoupled receptor are two distinct proteins that arefunctionally, but not structurally, linked together.Although these receptors contain seven membranespanning regions, they cannot directly alter intra-cellular activity upon binding of hormone. Instead,located at the cytoplasmic lining of the plasmalemmaand neighboring the receptor is a G protein thatstimulates cellular responses upon the formation ofhormone–receptor complexes. These G proteinsasotermed because they require GTP to functionacanbe either stimulatory (GS) or inhibitory (GI) in theiractions, with GS being far more commonplace.

All G proteins, whether GS or GI, consist of threesubunits termed α, β and γ. In the quiescent state,the α subunit is bound to guanosine diphosphate,but upon binding of hormone to its receptor, thisnucleotide is replaced by GTP, thus activating the Gprotein. Once activated, G proteins can stimulate a host of different intracellular second messengersystems, therefore regulatingaeven simultaneouslyanumerous intracellular activities. The major signaltransduction mechanisms linked with G proteinactivation will be addressed individually.1 Cyclic adenosine monophosphate (cAMP). In this second messenger system, activated G proteinsstimulate the enzyme adenylate cyclasealocated on the cytoplasmic lining of the plasmalemmaatoconvert ATP into cAMP. This reaction is similar tothe one stimulated by the interaction of hormonewith guanylate cyclase receptors where the bindingof hormone results in the conversion of GTP tocGMP. In addition to the nucleotide substrate used,the systems differ in that, unlike guanosine cyclase,the enzyme adenylate cyclase is not a constituent of the receptor molecule.

Newly formed cAMP is capable of triggering num-erous intracellular processes, primarily by activating

Page 38: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

22 chapter 2

various cAMP dependent protein kinases, also refer-red to as PKAs. As with all kinases, PKAs stimulatecellular responses by phosphorylating enzymes ofspecific biochemical pathways. Because each type oftarget cell expresses its own set of PKAs and kinaseactivated pathways, the same second messengersystem may be used to control biochemical res-ponses that are cell specific. And consistent withhormone action in general, at each step along theway the message of the hormone is amplified.

The production of cAMP occurs in a matter of sec-onds, consequently the cellular response is quicklyevident; unless hormone–receptor complexes arecontinually renewed the cellular action will be brief.Two enzymes terminate the actions induced bycAMP; phosphodiesterases cleave the bonds of cAMPtherefore inactivating it, and phosphatase activ-ity de-phosphorylates the enzymes stimulated byPKAs. As a result, responses typically evoked bycAMP are fast acting, and brief in duration.

However, not all biological responses associatedwith cAMP are of short duration. It is now knownthat cAMP can alter the transcription of certain pro-teinsamimicking the actions of steroids and thyroidhormonesaand therefore bring about long-termmodifications in cellular activity. Genes that are sensitive to cAMP regulation contain sequencesreferred to as cAMP response elements that act asenhancers of transcriptional activity when stimu-lated. This stimulation occurs when a specific typeof cAMP dependent protein kinase phosphorylatesthe cAMP responsive element binding protein (CREB).Once phosphorylated, these CREBs act as trans-cription factors and bind with the cAMP responseelements located on the DNA. The genes responsiveto these CREBs vary in accordance with target celltype. This, again, is an illustration of how differenttarget cells respond to the same signal transductionmechanism in a specific manner that is dictated bypre-existing ‘hard wiring’ of the cell.2 Phosphatidylinositolain another G protein relatedsecond messenger system, a phospholipid constitu-ent of the plasmalemmaaphosphatidylinositol—isdegraded by the activity of phospholipase C, a mem-brane bound enzyme that is stimulated by G proteinactivity. Upon binding of hormone to its membrane

receptor, phospholipase C cleaves phosphatidy-linositol into diacylglycerol (DAG) and inositoltriphosphate (IP3), each of which triggers a cellularresponse. Inositol triphosphate leaves the plasmal-emma to enter the cell’s cytoplasm where it re-acts with the endoplasmic reticulum to release its stored calcium into the cytosol. Increasing cytosoliccalcium levels is a common method of stimulatingvarious cellular activities via calcium sensitiveenzymes.

Unlike IP3, the newly produced DAG remainsbound to the cytoplasmic lining of the plasmalemmawhere it can activate the membrane bound proteinkinase C (PKC) enzyme. As its name suggests, PKCcan only be turned ‘on’ by DAG in the presence ofelevated cytosolic calcium levels, thus the actions of IP3 are synergistic to those of DAG. As with allkinases, PKC functions by phosphorylating, andthus activating, enzymes within the cell.

The activities stimulated by the phosphatidy-linositol signal transduction mechanism cease when IP3 is eliminated by its conversion to inositolthrough a process of de-phosphorylation, and DAGis inactivated by the addition of a phosphate group.The activities of intracellular enzymes stimulated byPKC are suppressed when cytosolic calcium levelsare returned to resting values.3 In addition to the direct coupling of ion channelopening with hormone binding as described earlier,receptor binding at the membrane may regulatechannel opening through a G protein intermediary.In this process, the membrane bound hormonereceptor and the channel that spans across the mem-brane are separate proteins. Upon formation of theligand-receptor complex, a nearby G protein locatedin the plasmalemma interacts with a neighboringchannel protein. This interaction causes a conforma-tional shift in the channel, resulting in its openingand movement of ions across the membrane.Typically, there is a large influx of ions into the cellresulting in a substantial increase in the cytosolicconcentration of that ion (Finn et al. 1996). Perhapsthe best example of this occurs in smooth musclecells where a sharp increase in cytosolic calcium levels occurs via G protein opening of ion speci-fic channels in the plasmalemma. At rest, cytosolic

Page 39: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

principles of endocrinology 23

calcium concentrations of these cells are in the range of 0.1–0.2 µmol·L–1, but upon hormone inducedstimulation of calcium channel linked G proteinsthese levels quickly rise to more than 1 mmol·L–1.This is a much greater increase than that observedfollowing the opening of ligand gated ion channels.

In some cases, newly available calcium does notby itself stimulate a biological response within theaffected cell. Instead, the greatly increased con-centration of calcium dramatically augments theprobability of its interaction with calcium specificintracellular binding proteins. Calmodulin is mostprominent among these binding proteins and itexists in almost all cells. This high affinity proteinpossesses four binding sites. When these sites areoccupied by newly available calciumawhich canalso originate from intracellular storesathe calcium-calmodulin complex activates enzymes, most com-monly kinases. In turn, these activate enzymes thatdirectly catalyze the cellular activities attributed to the actions of the hormone bound to the extracel-lular surface of the target. This cellular responsesubsides with the dissolution of hormone–receptorcomplexes, therefore closing ion channels. Adenosinetriphosphate-driven calcium pumps then delivercytosolic ions back to their intracellular (endoplas-mic reticulum) or extracellular sources of origin.

Integration of target cell responses to hormones

The integrative processes that characterize theendocrine system are not only evident during thesecretion of hormones but also in the responses oftarget tissue to those hormones. More specifically,the biological activity stimulated by one hormone in a target tissue can be modified by the action ofanother hormone. Such integrated responsivenessof target cells is best exemplified by the phenomenaof permissiveness, synergism, and antagonism.Synergism, also referred to as ‘potentiation’, occurswhen two different hormones stimulate the samebiological activity in the target cell. However, ratherthan the effects of the two hormones being additivein magnitude, the response is greater than would beevident by simply summing the responses demon-

strated when the cell responds to each hormoneindividually. To illustrate this, both growth hormoneand cortisol elicit lipolysis in adipocytes but, whenadministered simultaneously, the rate of triglycer-ide break down is actually much greater than itwould be if those hormones were given separatelyand their individual effects were added together.

In permissiveness the binding of one hormone atthe target cell must precede the binding of anotherhormone, which only then stimulates a biologicalresponse in the cell. In this case, it is said that the ini-tial hormone confers a permissive effect, allowingthe target cell to respond to the second hormone. Asan example of this, it has been found that in manytarget cells, the initial binding of thyroid hormoneenables epinephrine to exert its biological effects on those cells. Lastly, antagonism occurs when theinfluence of one hormone opposes, and effectivelyminimizes or even prevents, the action(s) of anotherhormone at the target cell. To exemplify this, growthhormone is known to antagonize the effects ofinsulin at their shared target tissues. As a result, the binding of growth hormone interferes with theability of insulin to promote glucose uptake andglycogen synthesis in the liver and skeletal muscle.

Concluding comments

Even in the brief description presented here, it isobvious that the mechanisms utilized by the endo-crine system to regulate target tissue responses arecharacterized by a high degree of complexity andintegration. Even within a single cell, physiologicalactivities are governed both by steroid and proteinhormones, while various intracellular signal trans-duction mechanisms are employed in an effort tomaintain homeostasis in the presence of continuousand diverse environmental perturbations. How-ever, it is now understood that many patholo-gical conditions, for example type 2 diabetes, can be directly attributed to dysfunction of these sig-nal transduction mechanisms. Accordingly, muchresearch is currently being conducted that willenhance our understanding of hormone regulatedsignal transduction in particular, as well as the func-tion of the endocrine system in general.

Page 40: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

24 chapter 2

References

Argetsinger, L.S., Campbell, G.S., Yang, X.et al. (1993) Identification of JAK2 as agrowth hormone receptor-associatedtyrosine kinase. Cell 74, 237–244.

Baulieu, E.E. (1990) Hormones, a complexcommunications network. In: Hormones:from Molecules to Disease (Baulieu, S.L. &Kelly, P.A., eds.). Chapman & Hall, NewYork: 3–171.

Bayliss, W.M. & Starling, E.H. (1902) Themechanism of pancreatic secretion.Journal of Physiology 28, 325–353.

Catelli, M.G., Binart, N., Jung-Testas, I. et al. (1985) The common 90-kD proteincomponent of non-transformed ‘8S’steroid receptors is a heat-shock protein.EMBO Journal 4, 3131–3135.

Chretian, M. & Seidah, N.G. (1981)Chemistry and biosynthesis ofpropiomelanocortin. Molecular andCellular Biochemistry 34, 101–127.

Dahlberg, E., Snochowski, M. &Gustafsson, J. (1981) Regulation of theandrogen and glucocorticoid receptorsin rat and mouse skeletal muscle cytosol.Endocrinology 108, 1431–1440.

Deschenes, M.R., Maresh, C.M.,Armstrong, L.E. et al. (1994) Enduranceand resistance exercise induce musclefiber type specific responses in androgenbinding capacity. Journal of SteroidBiochemical and Molecular Biology 50,175–179.

Finn, J.T., Greenwald, M.E. & Yau, K.W.(1996) Cyclic nucleotide-gated ionchannels: an extended family withdiverse functions. Annual Reviews inPhysiology 58, 395–426.

Firth, S.M. & Baxter, R.C. (2002) Cellularactions of the insulin-like growth factorbinding proteins. Endocrine Reviews 23,824– 854.

Goodman, H.M. (1994) Basic MedicalEndocrinology, 2nd edn. Raven Press,New York.

Guyton, A.C. & Hall, J.E. (1996) Textbook of Medical Physiology, 9th edn. W.B.Saunders, Co., Philadelphia.

Hedge, G.A., Colby, H.D. & Goodman,R.L. (1987) Clinical Endocrine Physiology.W.B. Saunders, Co., Philadelphia.

Heim, M.H. (1999) The Jak–STATpathway: cytokine signaling from thereceptor to the nucleus. Journal ofReceptor and Signal Transduction Research19, 75–120.

Illnerova, H., Sumova, A., Travnickova, Z.,Jac, M. & Jelinkova, D. (2000) Hormones,

subjective night and season of the year.Physiological Reviews 49 (suppl. 1),S1–S10.

Joab, I., Radanyi, C., Renoir, J.M. et al.(1984) Immunological evidence for acommon non hormone-bindingcomponent in ‘non-transformed’ chickoviduct receptors for four steroids.Nature 308, 850–853.

Kelly, R.B. (1985) Pathways of protein ineukaryotes. Science 230, 25–32.

Krieger, D.T., Liotta, A.S., Brownstein, M.J.& Zimmerman, E.A. (1980) ACTH, β-lipotropin and related peptides in brain,pituitary and blood. Recent Progress inHormone Research 36, 277–344.

Kronenberg, H., Melmed, S., Larsen, P.R. & Polonsky, K. (2003) Principles ofendocrinology. In: Williams Textbook ofEndocrinology (Larsen, P.R., Kronenberg,H., Melmed, S. & Polonsky, K., eds.).W.B. Saunders, Co., Philadelphia: 1–9.

Kuo, M.H. & Allis, C.D. (1998) Role ofhistone acetyltransferases anddeacetylases in gene regulation.Bioessays 20, 615–626.

Lazar, M.A. (2003) Mechanism of action ofhormones that act on nuclear receptors.In: Williams Textbook of Endocrinology(Larsen, P.R., Kronenberg, H., Melmed,S. & Polonsky, K., eds.). W.B. Saunders,Co., Philadelphia: 35–44.

Lewy, A.J., Sack, R.L., Miller, S. & Hoban,T.M. (1987) Anti-depressant andcircadian phase-shifting effects of light.Science 235, 352–354.

Liotta, A.S., Houghten, R. & Krieger, D.T.(1982) Identification of a β-endorphin-like peptide in cultured human placentalcells. Nature 295, 593–595.

McKenna, N.J., Lanz, R.B. & O’Malley,B.W. (1999) Nuclear receptorcoregulators: cellular and molecularbiology. Endocrine Reviews 20, 321–344.

Margioris, A.N., Liotta, A.S., Vaudry, N.,Boudin, G.W. & Krieger, D.T. (1982)Characterization of immunoreactivepropiomelanocortin-related peptides inrat testes. Endocrinology 113, 463–471.

Oichinik, M., Murray, T.F. & Moore, F.L.(1991) A corticosteroid receptor inneuronal membranes. Science 252,1848–1851.

Pedersen, K.B. & Vedickis, W.V. (2003)Quantification and glucocorticoidregulation of glucocorticoid receptortranscripts in two human leukemic celllines. Biochemistry 42, 10 978–10 990.

Pevet, P., Botherel, B., Slotten, H. &Saboureau, M. (2002) The chronobioticproperties of melatonin. Cell and TissueResearch 309, 183–191.

Porksen, N. (2002) The in vivo regulation ofpulsatile insulin secretion. Diabetologia45, 3–20.

Potier, M., Karl, M., Zhang, F. et al. (2002)Estrogen-related abnormalities inglomerulosclerosis-prone mice: reducedmesangial cell estrogen receptorexpression and prosclerotic response toestrogens. American Journal of Pathology160, 1877–1885.

Re, R. (2003) The intracrine hypothesis andintracellular peptide hormone action.Bioessays 25, 401–409.

Rhoades, R. & Pflanzer, R. (2003) HumanPhysiology, 4th edn. Thomson,Brooks/Cole, Pacific Grove, CA.

Scheidereit, C., Westphal, H.M., Carlson,C., Bosshard, H. & Beato, M. (1986)Molecular model of the interactionbetween the glucocorticoid receptor andthe regulatory elements of induciblegenes. DNA 5, 383–391.

Sherwood, L. (2004) Human Physiology:from Cells to Systems, 5th edn. Thomson,Brooks/Cole, Belmont, CA.

Short, R.V. (1985) Photoperiodism,melatonin and the pineal: it’s only amatter of time. In: Photoperiodism,Melatonin and the Pineal (Short, R.V., ed.).Pitman Press, London: 1–8.

Spiegel, A., Carter-Su, C. & Taylor, S.(2003) Mechanism of hormones that actat the cell surface. In: Williams Textbook ofEndocrinology (Larsen, P.R., Kronenberg,H., Melmed, S. & Polonsky, K., eds.).W.B. Saunders, Co., Philadelphia: 45–64.

Tamarkin, L., Baird, C.J. & Almeida, O.F.X.(1985) Melatonin: a coordinating signalfor mammalian reproduction. Science227, 714–720.

Tchaikovsky, V.S., Astratenkova, J.V. &Basharina, O.B. (1986) The effect ofexercises on the content and reception of the steroid hormones in rat skeletalmuscle. Journal of Steroid Biochemistry 24,251–253.

Vander, A., Sherman, J. & Luciano, D.(2001) Human Physiology: the Mechanismsof Body Function, 8th edn. McGraw-Hill,Boston.

Yakar, S., Wu, Y., Setser, J. & Rosen, C.J.(2002) The role of circulating IGF-I:lessons from human and animal models.Endocrine 19, 239–248.

Page 41: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

25

Scope of exercise testing in clinical medicine

An increasing number of investigators are usingmeasurements of human performance to probepathophysiology and disease mechanisms in a vari-ety of conditions. Figure 3.1 shows the exponentialincrease in the number of published, peer-reviewedrandomized trials that utilized clinical exercise test-ing. The widespread appeal of exercise testing isthat it allows clinicians to quantify physiologicalresponses in a controlled setting that better reflectsthe natural environment of their patients. Measure-ments of almost any physiological variable, rangingfrom cardiovascular to hormonal, made only at rest,

rarely predict the impact of disease on exerciseresponses.

Although tests like the 6-min walk (i.e. measuringthe distance walked by a subject in 6 min) continuesto be a useful assessment of integrated responses,such tools provide only crude insight into specificphysiological mechanisms of disease. How muchmore information we could have from these simpleprotocols if, for example, in addition to the distancewalked, we could measure body heat dynamics,mechanical work performed and shifts in intramus-cular water with minimally invasive and intrusivedevices.

Advances in the technology of exercise testing

Since the remarkable ‘growth spurt’ in human per-formance knowledge generated in the first half of the 20th century at such notable centers as theHarvard Fatigue Laboratory (Tipton 1998), thedevelopment of new, clinically useful, technologiesto assess human performance in response to physio-logical stresses like exercise has not kept pace withthe progress of many other areas of biomedicalinvestigation. Treadmills and cycle ergometers havechanged little since the first measurements of max-imal oxygen uptake were made in the 1920s. Thesedevices have proved to be very useful for testing theupper limits of gas exchange and metabolism, andare, therefore, suitable for studies of athletic perform-ance in which near maximal efforts are critical. Butthe emphasis on maximal efforts does not readilyreflect the level and type of physical activity that

Chapter 3

Exercise Testing: a Bridge Between theHigh-Tech and the Humanathe Need forInnovative Technologies

DAN M. COOPER

1966

–70

1971

–75

1976

–80

1981

–85

1986

–90

1991

–95

1996

–200

0

1600

1200

800

400

0

Nu

mb

er o

f Pu

bM

ed c

itat

ion

s

Fig. 3.1 Increasing number of clinical trials using exercisetesting. Using the PubMed search engine, it is clear thatthe utilization of exercise testing in clinical trials isincreasing rapidly.

Page 42: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

26 chapter 3

determines quality of life in the clinical setting.Moreover, typical exercise protocols are exhaustingand uncomfortable, and often inappropriate foryoung children, the elderly and persons with mostdiseases and disabilities (Cooper 1995; Metra et al.1998) (Fig. 3.2).

Gas exchange during exercise can be measuredprecisely, but only with cumbersome and some-times painful mouthpieces which, in and of them-selves, alter normal respiratory patterns (Lowhagenet al. 1999). Beyond the measurement of oxygen and carbon dioxide, little progress has been made to exploit the potentially rich insight into diseasemechanisms that could be gained from online, con-tinuous measurement of nitric oxide (NO) andvolatile organic compounds in the exhaled breath.Precise non-invasive quantification of physical activ-ity and energy expenditure under field conditions in free-living human beings remains an elusive goal; such tools, like the use of stable-isotopicallylabeled bicarbonate (Zanconato et al. 1992; Coggan et al. 1993), could revolutionize research focused onhealth outcomes in many areas of clinical and basicscience biomedical research.

Over the past 35 years, technological advanceshave facilitated an explosion of biomedical know-ledge, particularly in molecular biology and neuro-

science. Lagging behind this knowledge has beenthe development of tools for minimally invasivemeasures of neurological, intramuscular, cell sig-naling and vascular adjustments to exercise andother stresses that could be used easily and safely inhuman beings. Most troubling is that this lack ofprogress has occurred despite an increasing use oftraditional exercise testing to probe mechanisms of human disease and to develop new therapies.Moreover, there is growing recognition that healthimpairment directly related to physical inactivity isincreasing at an alarming rate (Booth et al. 2000;Cooper et al. 2004).

The clinical exercise testing technology gap hasoccurred despite a number of key technological andconceptual breakthroughs where basic engineeringresearch is pointing the way toward novel clinicalapplications. In the 1970s and 1980s, the notedHarvard biomedical engineer T. A. McMahon(McMahon 1984) reconfigured our understandingof the biomechanical principals that govern humanlocomotion. His theories were tested on a runningtrack built in his laboratory constructed so thatmechanical forces generated during human runningwere matched by forces generated within the track.Equally remarkable was that McMahon developeda paradigm in which the biomechanical mechanisms

Fig. 3.2 ‘Found art’ in the General Clinical Research Center (GCRC) Human Performance Laboratory. A 7-year-old girlcreatively expresses her thoughts on the Vo2max test. ‘I’m very sorry Dr. Cooper. I’m very tierd [sic] and I don’t want to dothis again. I’m so sorry.’

Page 43: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise testing 27

of human locomotion translated into theoreticalconstructs that involved biochemical, cardiovas-cular and neurological control mechanisms. Suchnovel uses of integrative physiology toward solvingbasic problems of human disease have proved to bea successful as a clinical research paradigm.

Imaging and spectroscopy approaches

A number of additional advances in non-invasiveimaging approaches towards clinical human exer-cise testing also need to be mentioned in this con-text. One involves attempts to link the tools ofremote imaging with physiological function dur-ing exercise. Anatomic imaging, such as can occurwith amazing precision using magnetic resonanceimaging, has been used to determine the profoundchanges in muscle water content and other physio-logical mechanisms that occur even with brief exercise. But Patten and colleagues in a recent com-prehensive review of T2 mapping of muscle noted:

Despite demonstration of the capacity forimaging phenomena relevant to both exercisephysiology and clinical diagnosis, to date therehave been surprisingly few clinical applicationsof T2 muscle mapping. Because MRI is nonin-vasive, it affords several advantages over tra-ditional diagnostic modalities such as musclebiopsy or EMG for diagnosis of metabolic andneuromuscular disorders in sports medicine,occupational medicine, and neurorehabilita-tion. MRI studies provide results rapidly forpurposes of diagnostic decision-making andalso offer outcome assessment for clinical orexercise interventions without the patient suf-fering the risks or discomforts associated withrepeated study (Patten et al. 2003).

In their review, Patten and coworkers went on topoint out that the alterations in T2 magnetic reson-ance imaging responses to exercise likely resultfrom two mechanisms: osmotically driven shifts ofmuscle water that increase the volume of the intra-cellular space; and from intracellular acidificationresulting from the end products of metabolism. Anexample of the use of these techniques in a pareticmuscle and in a control subject are shown in Fig. 3.3.

Remote biochemical imaging (e.g. 31P magneticresonance spectroscopy), pioneered by BrittonChance and his colleagues (Chance 1994), also holdsthe possibility of providing clinical investigatorswith real time measurements of intramuscular high-energy phosphate during human performance(Zanconato et al. 1993). For example, Scheuermann-Freestone et al. (2003) recently examined both cardiac and skeletal muscle energetics during exer-cise in patients with type 2 diabetes (Fig. 3.4).Remarkably, although their cardiac morphology,mass and function appeared to be normal, thepatients with diabetes had significantly lower phos-phocreatine/adenosine triphosphate (PCr/ATP)ratios than the healthy volunteers. The cardiacPCr/ATP ratios correlated negatively with the fast-ing plasma free fatty acid concentrations. Althoughskeletal muscle energetics and pH were normal atrest, PCr loss and pH decrease were significantlyfaster during exercise in the patients with diabetes,who had lower exercise tolerance, and PCr recoverywas slower in the patients. These investigators con-cluded that type 2 diabetic patients with apparentlynormal cardiac function have impaired myocardialand skeletal muscle energy metabolism related tochanges in circulating metabolic substrates.

A major breakthrough in the understanding ofexercise responses and their alteration in diseasestates resulted from yet another interaction betweenengineers and physicians. The development ofbreath-by-breath measurement of gas exchange was pioneered in large measure in the Harbor–University of California, Los Angeles (UCLA)Laboratory of Drs. Brian Whipp and KarlmanWasserman (Wasserman et al. 1973). Wasserman, anMD with a PhD in physiology, was a fellow in thenoted Cardiovascular Research Institute (CVRI) atthe University of California, San Francisco in the1960s. Whipp and Wasserman collaborated primar-ily with two engineers, Dr. William Beaver and, sub-sequently, Dr. Norman Lamarra. Lamarra receivedhis PhD at UCLA in aerospace engineering for adoctoral thesis devoted, oddly enough, entirely toanalyzing the ontransient kinetics of oxygen uptakeduring exercise in human beings (Lamarra 1982)!Investigations into how disease influences thesekinetics, and how these measurements prove to be

Page 44: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

28 chapter 3

Fig. 3.3 T2-weighted axial magnetic resonance images of the arm obtained using a multiecho sequence (TE 20, 40, 60, 80ms, TR = 2000, I NEX, 18 cm FOV) with in-plane resolution of 0.6–0.8 mm. The upper panel images illustrate the paretic(left) and non-paretic (right) arms at rest in an adult with post-stroke hemiparesis of 24 months duration. The lower panelsillustrate the same arms following 40 dynamic elbow flexions performed at 80% of maximal effort. Following exercise,increased signal intensity (T2) is demonstrated in the flexor compartment bilaterally. The paretic side demonstratesmarkedly less activity-dependent increase in T2 and documents impaired muscle activation associated with post-strokehemiplegia. (Data from Patten et al. 2003.)

Page 45: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise testing 29

useful indicators of disease and its therapy, haveproceeded at a relatively slow pace.

The pioneers in the field of exercise physiologyclearly recognized that the biological responses to thestress of exercise could ultimately be used to gain agreater understanding of fundamental processes atthe cellular and subcellular level. Wasserman’s 1975cartoon of ‘gears’ linking cellular function to gasexchange measured at the mouth aptly illustratesthis concept (Fig. 3.5). But the primary use of exer-cise testing in the clinical setting has remainedalmost invariably focused on cardiovascular meas-urements. As noted by Wasserman and colleagues:

The authors would like to dispel a concept thathas developed in medicine, i.e., that there is acardiac stress testing and pulmonary stresstesting. It is impossible to stress only the heartor only the lungs. Exercise requires the co-

ordinated function of the heart, the lungs, andthe peripheral and the pulmonary circulation tomatch the increased cellular respiration requiredto live and work (Wasserman et al. 1987).

We would add only that the ‘co-ordinated function’includes the neuromuscular and cellular signaltransduction adaptive mechanisms as well.

Development of new technologies—robots, strokes and maturation of motor control

In some cases, existing technologies are simply not sufficient to test exercise and motor control incertain individuals and new devices need to bedeveloped. For example, in dealing with stroke vic-tims, traditional treadmills or cycle ergometers areoften not feasible as either diagnostic or therapeutic

Pi

5 0 –5 –10 –15p.p.m.

PDE

ATP

PCr Controlresting

pH = 7.07

Diabeticresting

pH = 7.11

Diabeticend-exercise

at 5.1 minpH = 6.57

Control after5.1 minexercise

pH = 7.00

γ α β

10 5 0 –5 –10 –15 –20p.p.m.

10 5 0 –5 –10 –15 –20p.p.m.

5 0 –5 –10 –15p.p.m.

Fig. 3.4 Typical calf muscle 31Pmagnetic resonance spectra from acontrol subject and a patient withtype 2 diabetes at rest (upper panel,number of scans = 64), from the samepatient at the end of exercise, and the same matched control at theequivalent time (5.1 min) of exercise(lower panel, number of scans = 16).PCr, phosphocreatine; PDE,phosphodiesters; Pi, inorganicphosphate; α, β and γ indicate thethree phosphate groups of adenosinetriphosphate (ATP). Cytosolic pHwas calculated from the chemicalshift of Pi relative to PCr. Abscissashows chemical shift in parts permillion (p.p.m.). (Data fromSheuermann-Freestone et al. 2003.)

Page 46: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

30 chapter 3

tools because of the constraints and limitations tomotor control caused by the brain injury. To getaround these limitations, mechatronic devices andhaptic robots have been developed by a number ofinvestigators. Using these tools, new insight into themechanisms that govern motor activity in strokevictims (Lum et al. 2002).

For example, recent evidence suggests that braininjury can impair the ability to independently activ-ate shoulder and elbow muscles. Reinkensmeyer et al. (2002) hypothesized that if muscle activationpatterns are constrained, then brain-injured sub-jects should not be able to accurately grade initialhand movement direction during reaching toward a broad range of target directions. To test this hypo-thesis, Reinkensmeyer et al. used the mechatronichaptic robot to measure hand trajectories duringreaching in three-space by 16 subjects with hemi-paretic stroke to an array of 75 targets distributedthroughout the workspace (Fig. 3.6).

Results of these innovative studies clearly suggestthat there are two identifiable classes of directionalcontrol following stroke: largely preserved andseverely constrained. Since it has been hypothesizedthat lower pathways substitute for corticospinalones following stroke, a possible explanation for

CO2 FLOW

O2 FLOW

O2 CONSUM

CO2 PROD

INSPIRED

EXPIRED

Muscleactivity

PeripheralcirculationMitochondrion

Physiological response:

Muscle LungsHeartblood

Creat PO4

Pyr Lac

Pulmonarycirculation

O2 and CO2transport

Ventilation(VA + VO = VE)

QCO2

QCO2 Dilate RecruitSV

VCO2

VO2QO2 R

V

LV

VT

QO2 Recruit DilateHR f

Fig. 3.5 The metabolic ‘gears’ thatlink ventilation during exercise to the cellular and subcellular level.(From Wasserman et al. 1987.)

10 cm(a)

(b)

θ

Fig. 3.6 Example hand trajectories for the ipsilesional (a)and contralesional arms (b) of a subject with severe stroke.The contralesional (left) arm trajectories are flipped aboutthe sagittal plane. Reaching direction (θ) is definedpositive for movements to the right of straight ahead.Note that movement was constrained to essentially twodirections (medial and lateral) for the severely impairedsubject, even though substantial active range of motionwas preserved in these directions. (Data fromReinkensmeyer et al. 2002.)

Page 47: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise testing 31

these two classes is that directional control is largelypreserved if some threshold fraction of corticospinalpathways is spared. This hypothesis should betestable in the future via detailed functional imag-ing of key neural tracts.

The use of robots in measuring motor control isnot limited to brain-injured patients. Interestingly, achance interaction between Reinkensmeyer’s groupand the author of this chapter led to the idea thatmechatronic haptic robots could be used to measuremotor control during development in normal chil-dren (Fig. 3.7). Children do not typically appear tomove with the same skill and dexterity as adults,although they can still improve their motor perform-ance in specific tasks with practice.

One possible explanation is that their motor per-formance is limited by an inherently higher level ofmovement variability, but that their motor adaptiveability is robust to this variability. To test this hypo-thesis, Takahashi et al. (2003) examined motor adap-tation of 43 children (aged 6–17 years) and 12 adultsas they reached while holding the tip of a lightweightrobot. The robot applied either a predictable, velocity-dependent field (the ‘mean field’) or a similar fieldthat incorporated stochastic variation (the ‘noisefield’), thereby further enhancing the variability ofthe subjects’ movements. Children exhibited greaterinitial trial-to-trial variability in their unperturbedmovements, but were still able to adapt comparablyto adults in both the mean and noise fields. Further-more, the youngest children (aged 6–8 years) wereable to reduce their variability with practice to levelscomparable to the remaining children groups, thoughnot as low as adults. These results indicate that chil-dren as young as 6 years possess adult-like neuralsystems for motor adaptation and internal modelformation that allow them to adapt to novel dynamicenvironments as well as adults on average despiteincreased neuromotor or environmental noise.

Performance following adaptation is still morevariable than adults, however, indicating that move-ment inconsistency, not motor adaptation inability,ultimately limits motor performance by childrenand may thus account for their appearance of inco-ordination and more frequent motor accidents (e.g.spilling, tripping). The results of this study also sug-gest that movement variability in young children

may arise from two sourcesaa relatively constant,intrinsic source related to fundamental physiolo-gical constraints of the developing motor system, anda more rapidly modifiable source that is modulateddepending on the current motor context.

Exhaled nitric oxide and exercise

Clinicians are increasingly questioning the value of traditional measures of pulmonary function suchas the forced expiratory volume in 1 s (FEV1) socommonly used to assess childhood asthma (Spahnet al. 2004). A growing body of research has focusedon arguably more direct measurements of lunginflammation, such as exhaled NO (Paredi et al.2002). NO performs many important functions inthe lungs and can be detected in the exhaled breathof humans. Inflammatory diseases such as asthmaand cystic fibrosis alter exhaled NO levels. This hasgenerated interest in utilizing exhaled NO as a non-invasive marker of lung inflammation.

However, the exchange dynamics of NO aremarkedly different from the respiratory gases (oxy-gen and carbon dioxide) whose exchange occurspredominantly in the alveolar region. In contrast,NO exchange occurs in both alveolar and airwaycompartments, and is thus highly dependent on theexhalation flow rate. This feature of NO exchangehas confounded interpretation of exhaled NO in avariety of clinical and physiological settings. Giventhe nature of NO exchange dynamics, and the multi-system physiological responses to exercise, it is notsurprising that there are inconsistencies in thereports of the impact of exercise on exhaled NO.

Recently, a number of investigators have devel-oped paradigms to distinguish alveolar and air-way contributions to exhaled NO (George 2004)(Fig. 3.8). This approach provides greater specificitythan exhaled concentration alone, and thus may be able to address several unresolved questionsregarding the impact of exercise on NO exchange.Given the nature of NO exchange dynamics, and themultisystem physiological responses to exercise, itis not surprising that there are inconsistencies in the reports of the impact of exercise on exhaled NO. After exercise, exhaled NO concentration hasbeen reported to be increased (Bauer et al. 1994),

Page 48: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

32 chapter 3

1.5

1.0

0.5

0

Reach time (s)

6–8 9–12 13–17

Trials(a)

> 17

1.5

1.0

0.5

011–20 Last 10 Noise+

Trials(b)

Spatial variability (cm)

1.0

0.8

0.6

0.4

0.2

Temporal variability (s)

Trials(c)

100

80

60

40

20

011–20 Last 10 Noise+

Trials(d)

Scoring rate (%)

011–20 Last 10 Noise+

* ***

6–89–1213–17> 17

Fig. 3.7 Motor performance measures. (a) Reach time depended on age grouping (analysis of variance [ANOVA] linearcontrast, p < 0.001). (b) Spatial variability depended on age grouping early in the experiment (mean over trials 11–20,ANOVA linear contrast over children groups, p < 0.001) but became independent of age grouping (p = 0.99) by the end ofthe experiment (i.e. last 10 trials). Children aged 6–8 years significantly reduced their spatial variability (paired one-sidedt-test; ** p < 0.01) by the end of the experiment, but adults still maintained significantly lower levels compared to all otherchildren groups (ANOVA planned comparison, p = 0.002). Spatial variability was significantly increased by the end of thenoise field (‘noise+’ indicates last 10 trials of noise field) for all age groups (paired one-sided t-test, p < 0.001 all age groups)to levels that did not depend on age grouping (ANOVA linear contrast, p = 0.39). (c) Temporal variability also dependedon age grouping over trials 11–20 (ANOVA linear contrast over children groups, p < 0.001) but became independent of age grouping by the last 10 trials of experiment (p = 0.38). Children aged 6–8 years significantly reduced their temporalvariability (paired one-sided t-test, * p ≤ 0.05) by the end of the experiment. (d) Children aged 6–8 years improved theirtiming score rate (percent ‘just right’, t-test, *p ≤ 0.05) by the end of the experiment. Error bars show standard deviationacross subjects. (Data from Takahashi et al. 2003.)

Page 49: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise testing 33

unchanged (Iwamoto et al. 1994), or decreased(Maroun et al. 1995). By distinguishing alveolar andairway contributions to exhaled NO, Shin et al.(2003) recently used a two-compartmental model of NO exchange during exercise in an attempt toprovide greater specificity than exhaled concentra-tion alone. They hoped to able to address severalunresolved questions regarding the impact of exer-cise on NO exchange.

Significant changes were observed in J’awNO,DawNO and CawNO 3-min post-exercise challenge,despite no significant changes in exhaled con-centration (CNOplat). DawNO (mean ± SD) increased (37.1 ± 44.4%), whereas J’awNO and CawNO decreased(−7.27 ± 11.1%, −26.1 ± 24.6%, respectively) 3-minpost-exercise. Shin et al. (2003) concluded that theflow-independent NO parameters provide greaterspecificity in characterizing NO exchange. It seemsthat exercise acutely enhances elimination of NOfrom airway tissue stores. This effect may be due toenhanced ventilation or an enhanced ability of NOto diffuse from the airway tissue to the gas phase.

The latter suggests endogenously produced NOmay be useful to probe metabolic and structural fea-tures of the airways during exercise.

Linking exercise testing to the new biology

The new technologies of genomic profiling, pro-teomics and flow cytometry have opened novelvenues of research for exercise physiologists. Therecent discoveries of the impact of physical activityon stress, inflammatory and immune function(Fleshner et al. 2003; Pedersen et al. 2003; Shephard2003) have created a paradigm shift in our ability to understand the link between physical activityand health. For example, Fehrenbach et al. (2000)recently examined the role of exercise and fitness onleukocyte expression of key immune modulators,the heat shock proteins (HSPs) (Fig. 3.9). They foundlarge increases in certain HSPs within the leuko-cytes of subjects following exercise. HSPs inhibitnuclear factor-κB, and this may explain the HSP car-dioprotective effect that has been noted previously( Joyeux et al. 1999; Powers et al. 2002).

Summary

Investigators are increasingly requesting humanperformance/exercise studies on populations inwhom the technology of ‘traditional’ exercise test-ing is inadequate; namely, the elderly, children andthe disabled. The challenge we now face is to buildon the past century of progress in exercise physi-ology by investing in new technologies and approachthat can help us understand how exercise is linkedto fundamental disease processes. Moreover, thenew approaches and technologies must be used forintegrating exercise testing with innovative, multi-disciplinary, research tools in biology that can pro-vide new insights into mechanisms of disease at thesystemic and cellular level.

Acknowledgement

This work was supported in part by NationalInstitutes of Health Grant HD26939 and the UCISatellite GCRC MO1 RR00827.

Airway region(compartment #2)

Alveolar region(compartment #1)

DawNO*Cair

JawNO = JawNO – DawNOCair

= DawNO (CawNO – Cair)

Cexh(t)Calv, ss

JawNO

Fig. 3.8 Schematic of two-compartment model used todescribe nitric oxide (NO) exchange dynamics. ExhaledNO concentration (CEno) is the sum of two contributions,the alveolar region and the airway region, which dependson three flow-independent parameters: maximum totalvolumetric flux of NO from the airway wall (JawNO, pls),diffusing capacity of NO in the airways (DawNO, pls

1,p.p.b.1), and steady-state alveolar concentration (CAno,p.p.b.). JawNO, total flux (pls) of NO between the tissue andgas phase in the airway and is an inverse function of theexhalation flow rate (VE); Cno, concentration of NO in thegas phase within the airway compartment. (From Georgeet al. 2004.)

Page 50: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

34 chapter 3

References

Bauer, J.A., Wald, J.A., Doran, S. & Soda,D. (1994) Endogenous nitric oxide inexpired air: effects of acute exercise inhumans. Life Sciences 55, 1903–1909.

Booth, F.W., Gordon, S.E., Carlson, C.J. &Hamilton, M.T. (2000) Waging war onmodern chronic diseases: primaryprevention through exercise biology.Journal of Applied Physiology 88, 774–787.

Chance, B. (1994) Non-invasiveapproaches to tissue bioenergetics.Biochemical Society Transactions 22,983–987.

Coggan, A.R., Habash, D.L., Mendenhall,L.A., Swanson, S.C. & Kien, C.L. (1993)Isotopic estimation of CO2 productionduring exercise before and afterendurance training. Journal of AppliedPhysiology 75, 70–75.

Cooper, D.M. (1995) Rethinking exercisetesting in children: a challenge. AmericanJournal of Respiratory and Critical Care 152,1154–1157.

Cooper, D.M., Nemet, D. & Galassetti, P.(2004) Exercise, stress, and inflammation

HSP

27—

mR

NA

1.5

1.0

0.5

0.0

* *

RestTime

+0 h +3 h +24 h

HSP

70—

mR

NA

1.5

1.0

0.5

0.0Rest

Time(a) (b)+0 h +3 h +24 h

Fig. 3.9 Descriptional presentation of mRNA expression of heat shock proteins HSP27 (a) and HSP70 (b) in leukocytes of athletes at rest and immediately, 3 h and 24 h after the half-marathon (n = 12). Semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) was performed to assess mRNA expression. HSP27, HSP70 and β-actin wereamplified under conditions to allow relative comparisons for a given mRNA. The specific mRNA values are described inrelative units normalized to transcript levels of β-actin. Each curve represents a single subject. *Significant changescompared with resting values (P < 0.05). (Data from Fehrenbach et al. 2000.)

in the growing child: from the bench tothe playground. Current Opinions inPediatrics 16(3), 286–292.

Fehrenbach, E., Niess, A.M., Schlotz, E. et al. (2000) Transcriptional andtranslational regulation of heat shockproteins in leukocytes of endurancerunners. Journal of Applied Physiology89(2), 704–710.

Fleshner, M., Campisi, J. & Johnson, J.D.(2003) Can exercise stress facilitateinnate immunity? A functional role forstress-induced extracellular Hsp72.Exercise Immunology Review 9, 6–24.

George, S.C., Hogman, M., Permutt, S. &Silkoff, P.E. (2004) Modeling pulmonarynitric oxide exchange. Journal of AppliedPhysiology 96, 831–839.

Iwamoto, J., Pendergast, D.R., Suzuki, H. & Krasney, J.A. (1994) Effect of gradedexercise on nitric oxide in expired air in humans. Respiration Physiology 97,333–345.

Joyeux, M., Godin-Ribuot, D., Yellon,D.M., Demenge, P. & Ribuot, C. (1999)Heat stress response and myocardial

protection. Fundamental and ClinicalPharmacology 13, 1–10.

Lamarra, N. (1982) Ventilatory control,cardiac output, and gas exchange dynamicsduring exercise transients in man. PhDthesis. UCLA, Los Angeles.

Lowhagen, O., Arvidsson, M., Bjarneman,P. & Jorgensen, N. (1999) Exercise-induced respiratory symptoms are notalways asthma. Respiratory Medicine 93,734–738.

Lum, P., Reinkensmeyer, D., Mahoney, R.,Rymer, W.Z. & Burgar, C. (2002) Roboticdevices for movement therapy afterstroke: current status and challenges to clinical acceptance. Topics in StrokeRehabilitation 8, 40–53.

McMahon, T.A. (1984) Muscles, Reflexes,and Locomotion. Princeton UniversityPress, Princeton, NJ.

Maroun, M.J., Mehta, S., Turcotte, R.,Cosio, M.G. & Hussain, S.N. (1995)Effects of physical conditioning onendogenous nitric oxide output duringexercise. Journal of Applied Physiology 79,1219–1225.

Page 51: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise testing 35

Metra, M., Nodari, S., Raccagni, D. et al.(1998) Maximal and submaximalexercise testing in heart failure. Journal ofCardiovascular Pharmacology 32 (suppl. 1),S36–S45.

Paredi, P., Kharitonov, S.A. & Barnes, P.J. (2002) Analysis of expired air foroxidation products. American Journal ofRespiratory and Critical Care Medicine 166,S31–S37.

Patten, C., Meyer, R.A. & Fleckenstein, J.L.(2003) T2 mapping of muscle. Seminars inMusculoskeletal Radiology 7, 297–305.

Pedersen, B.K., Steensberg, A., Keller, P. et al. (2003) Muscle-derived interleukin-6: lipolytic, anti-inflammatory andimmune regulatory effects. PflugersArchiv 446, 9–16.

Powers, S.K., Lennon, S.L., Quindry, J. & Mehta, J.L. (2002) Exercise andcardioprotection. Current Opinion inCardiology 17, 495–502.

Reinkensmeyer, D.J., McKenna, C.A.,Kahn, L.E. & Kamper, D.G. (2002)Directional control of reaching ispreserved following mild/moderatestroke and stochastically constrained

following severe stroke. ExperimentalBrain Research 143, 525–530.

Scheuermann-Freestone, M., Madsen, P.L.,Manners, D. et al. (2003) Abnormalcardiac and skeletal muscle energymetabolism in patients with type 2diabetes. Circulation 107, 3040–3046.

Shephard, R.J. (2003) Adhesion molecules,catecholamines and leucocyteredistribution during and followingexercise. Reviews on Environmental Health33, 261–284.

Shin, H.W., Rose-Gottron, C.M., Cooper,D.M., Hill, M. & George, S.C. (2003)Impact of high-intensity exercise onnitric oxide exchange in healthy adults.Medicine and Science in Sports and Exercise35, 995–1003.

Spahn, J.D., Cherniack, R., Paull, K. &Gelfand, E.W. (2004) Is forced expiratoryvolume in 1 second the best measure ofseverity in childhood asthma? AmericanJournal Respiratory and Critical CareMedicine 169, 784–786.

Takahashi, C.D., Nemet, D., Rose-Gottron,C.M. et al. (2003) Neuromotor noiselimits motor performance, but not motor

adaptation, in children. Journal ofNeurophysiology 90, 703–711.

Tipton, C.M. (1998) Contemporary exercise physiology: fifty years after theclosure of Harvard Fatigue Laboratory.Exercises and Sport Science Review 26,315–339.

Wasserman, K., Whipp, B.J., Koyal, S.N. &Beaver, W.L. (1973) Anaerobic thresholdand respiratory gas exchange duringexercise. Journal of Applied Physiology 35,236–243.

Wasserman, K., Hansen, J.E., Sue, D.Y. &Whipp, B.J. (1987) Principles of ExerciseTesting and Interpretation. Lea & Febiger,Philadelphia, PA.

Zanconato, S., Cooper, D.M., Barstow, T.J.& Landaw, E. (1992) 13CO2 washoutdynamics during intermittent exercise in children and adults. Journal of AppliedPhysiology 73, 2476–2482.

Zanconato, S., Buchthal, S., Barstow, T.J. & Cooper, D.M. (1993) 31P-magneticresonance spectroscopy of leg musclemetabolism during exercise in childrenand adults. Journal of Applied Physiology74, 2214–2218.

Page 52: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

36

Introduction

Methods to quantify peptide hormone concentra-tions in biological fluids are a basic requirement formedical practice and biomedical research in thefield of endocrinology. Such methods ideally shouldbe very specific, identifying precisely and quantify-ing accurately only the hormone of interest withoutinterference from closely related peptides or un-related matrix components. They must be extremelysensitive, allowing the detection of peptide hor-mones at the low concentrations observed in physio-logical and pathophysiological situations. Usually,peptide hormone concentrations in circulation arein the nano- or picomolar range, making the sensiti-vity of the methods a crucial issue. Finally, measure-ment methods should be easy and rapid, allowingapplication to large series of samples in an appropri-ate time. These methodological aspects of hormonemeasurement become even more important whenthe analyses are done in the framework of the fightagainst doping in sports. Because of the potentiallysevere consequences for the athlete, the commercialand political interests involved and the complicatedethical and legal background, the level of precision,specificity and reliability for hormone measurementsmust be extremely high in this field. For example,the cross-reactivity from specific molecular isoformsof peptide hormones in immunoassays is rarelyknown for commercial assays frequently used inclinical practice, whereas this is a crucial point forseveral doping tests described below (chorinonicgonadotropin [hCG], erythropoietin [EPO], humangrowth hormone [hGH]). Finally, large-scale anal-

ysis of samples in an extremely short time period isoften required during competitive events, enforcingthe need for rapid analytical methods.

During the last 30 years, enormous progress hasbeen made in the attempt to develop analyticalmethods for peptide hormone quantification fulfill-ing the above mentioned criteria. However, fromthe early days of radioimmunoassays to the latestdevelopments in mass spectrometry (MS) tech-niques, it remains crucial to be aware of the pitfallseach method includes to avoid misinterpretation of the data generated. In some cases, it might beimpossible to combine different goals by onemethod: For example, there is no doubt that MS is aunique tool to identify a molecule with an extremelyhigh degree of certainty (Binz et al. 2003; Gam et al.2003; Kast et al. 2003). However, today’s MS methodsstill are complex, time consuming and in many casesrequire a very sophisticated sample preparation.The progress made in the applicability of MS tech-nologies to analyze purified peptide hormone pre-parations is not paralleled by the same progress in analyzing hormone mixtures in more complexbiological matrices like serum (Liu & Bowers 1997;Black & Bowers 2000; Wu, S.L. et al. 2002), and thehigh degree of certainty achieved under optimalconditions by no means indicates that MS would notbe susceptible to interferences (Annesley 2003).Finally, the costs of the equipment necessary are stillmuch higher than those for immunological hor-mone measurement methods. The rapid develop-ment of new techniques in the area of MS togetherwith the development of instruments designed forhigh throughput analyses, the introduction of

Chapter 4

Measurement of Peptide Hormones

MARTIN BIDLINGMAIER, ZIDA WU AND CHRISTIAN J. STRASBURGER

Page 53: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

measurement of peptide hormones 37

automated systems for sample preparation and theincrease in sensitivity recently achieved by researchgroups might change the picture in the future (forreview see Binz et al. 2003). However, until today the vast majority of hormone analyses in clinicalpractice and research is still done by the ‘traditional’immunoassay techniques. Thus, this article isfocused on the pitfalls and potentials of peptide hor-mone measurements by immunoassay techniques.

General aspects of immunoassaymethods

Immunoassays employ the unique capability ofspecific immunoglobulins (antibodies) to recognizeand bind a certain three-dimensional structure onthe surface of a molecule (the so-called ‘epitope’).This antigen–antibody reaction provides a highdegree of specificity. In combination with the use of powerful labeling substances for antibodies orhormones, the dynamic range of immunoassayscould be greatly improved. Traditionally, radioactiv-ity had been used as a detection system in hormoneassays. Driven by concerns regarding environmen-tal, economical and health aspects of radiation, different non-isotopic methods have been devel-oped for signal detection: These newer systemsemploy either an enzyme-catalyzed colorimetric or chemiluminescence reaction oraalternativelyaafluorescence dye for signal detection. Among theadvantages of these non-isotopic methods is the stability of the labelaradioactive labels undergo a decay, making lot-to-lot differences a commonproblem. In some cases, the sensitivity of modernnon-isotopic detection systems exceeds that ofradioactive labels, making their use even more popular. However, in general the sensitivity of animmunoassay method primarily depends on theaffinity of the specific antibodies used, and often thedetection label is chosen simply because a certainmeasurement device is present in a laboratory.

Technical aspects of immunoassaytechnology and its implications

From a methodological point of view, immunoas-says can be divided in two major subtypes: the

classical ‘competitive’ immunoassay and the ‘sand-wich type’ immunoassay (Figs 4.1 and 4.2). In acompetitive immunoassay, the hormone present ina sample ‘competes’ for antibody binding with alabeled form of the same hormone added to thesample. The higher the endogenous hormone con-centration in a sample is, the lower the probability is that a labeled hormone molecule (called ‘tracer’)binds to the antibody. Thus, the hormone concen-tration in the sample is inversely correlated to thesignal obtained in the assay. Depending on the labelused, competitive assays have been named radio-immunoassay (RIA), enzyme-immunoassay (EIA),luminescence-immunoassay (LIA) or fluorescence-immunoassay (FIA). In contrast, ‘sandwich type’immunoassays consist of a ‘capture antibody’, whichbinds the hormone from the sample, and a labeled‘detection antibody’, which is directed againstanother epitope on the hormone surface and thusallows ‘staining’ of the hormone molecules boundby the capture antibody. The more the hormone ispresent in a sample, the more the hormone is boundby the capture antibody, which in turn is trans-lated into a signal by the ‘detection antibody’. Thus,the signal obtained is directly proportional to theamount of hormone present in the sample. Similarto the above mentioned nomenclature for the com-petitive assays, the names of sandwich type immuno-assays are related to the detection system used:radioimmunometric assay (IRMA), enzyme linkedimmunosorbent assay (ELISA), luminometric assay(ILMA) or fluorometric assay (IFMA).

The difference in assay design between competit-ive and sandwich type immunoassays has majorimplications on their applications and on the inter-pretation of the results: whereas competitive assaysrequire only one antibody andacorrespondinglya

one epitope, sandwich type immunoassays requiretwo different antibodies and thus two different epitopes. These epitopes must be in spatial distance,because otherwise binding of the capture antibodywould interfere with the binding of the detectionantibody. Accordingly, sandwich type immuno-assays require a ‘larger molecule’ and therefore are used to measure larger peptide hormones likeinsulin, growth hormone (GH) or EPO, whereas the smaller peptides (adrenocorticotropic hormone

Page 54: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

38 chapter 4

Signal

0.0(b)(a)

1.0Hormone (ng·mL–1)

10.0 100.0

1.0

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

0.0

Fig. 4.1 (a) Competitive assayaprinciple. Labeled hormone (‘tracer‘) and hormone from the sample compete for bindingto antibody. (b) Competitive assayacalibration curve. The higher the hormone concentration, the lower the probabilitythat a labeled tracer molecule is bound. Consequence: the more hormone, the lower the signal.

Signal

0.1(b)(a)

1.0Hormone (ng·mL–1)

10.0 100.0

1 000 000

100 000

10 000

1000

Fig. 4.2 (a) Sandwich assayaprinciple. (i) Hormone from the sample binds to capture antibody. (ii) Bound hormone isdetected by labeled antibody. (b) Sandwich assayacalibration curve. The higher the hormone concentration, the higherthe probability that a labeled detection antibody is bound. Consequence: the more hormone, the more signal.

Page 55: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

measurement of peptide hormones 39

[ACTH], corticotropin-releasing hormone [CRH],growth hormone releasing hormone [GHRH]) andsteroid hormones frequently are measured by com-petitive assays. The advantage of the sandwich typeassay format is that the recognition of a hormonemolecule requires the presence of two independentepitopes, making the methodaat least theoreticallyamore specific and less prone to cross-reactivity ofpartially related peptides. In addition, the analyte(the hormone to be measured) and the calibrator(the standard preparation used) are chemicallyidentical, whereas in the case of competitive assays,the calibrator is a modified, labeled hormone. Thesemodifications can lead to differences in the affinityof the antibody to the naturally occurring hormonein a sample and to the modified hormone used asthe calibrator of the assay.

Standardization of immunoassays

It is important to mention that all immunoassaymethods are ‘relative’ in nature. This means, that the concentration of the substance of interest in asample is determined in comparison to the concen-tration contained in the calibrator. More precisely,the concentration is determined by comparing theability of the antibodies used to bind the hormone in a biological sample (e.g. a patient’s serum) and inthe calibrator sample, respectively. Theoretically,this measurement technique relies on three funda-mental assumptions: (i) the calibrator is identical tothe substance of interest in its physicochemicalproperties and in its three-dimensional structure;(ii) the epitopes on the surface of the molecule ofinterest are freely accessible for the antibody in boththe biological sample and in the calibrator; (iii) thematrix of the calibrator is identical to the matrix ofthe sample. Obviously it is very difficult to realizethese assumptions in an assay for peptide hormones:In many cases, no international reference prepara-tion (IRP) is available, whereas in other cases morethan one reference preparation exists. In the case ofhGH, the IRP 80/505 is of pituitary extraction,whereas the IRP 88/624 and IRP 98/574 are ofrecombinant origin. In addition, some of the refer-ence preparations are of very poor quality andimpure (e.g. the IRP for insulin-like growth factor I

[IGF-I] contains only about 40% IGF-I [Quarmby et al. 1998]). To make the situation more complic-ated, many peptide hormones naturally occurringin the human body are a mixture of molecular isoforms rather than a homogenous substance(Nagy et al. 1994). This has been documented in verymuch detail, especially for hCG (Birken et al. 2003;Lottersberger et al. 2003), but also for hGH(Baumann 1999; Boguszewski 2003). Depending onthe specific question an investigator or clinicianattempts to answer by measuring a hormone’s con-centration, the answer which of these isoforms orwhich mixture of isoforms would be the suitable‘reference’ preparation varies significantly. In 1991,Roger Ekins, one of the inventors of immunoassaytechnology wrote that standardization of immuno-assays for heterogeneous antigens is impossible(Ekins 1991), and the ongoing discussions on assaystandardization clearly demonstrate that we are farfrom having reached a consensus (Roger & Lalhou1994; Quarmby et al. 1998; Ranke et al. 2001). As tothe second assumption, one has to be aware that for many peptide hormones high- or low-affinity‘binding proteins’ naturally occur in circulation(Baumann et al. 1988). Present in a sample, thesebinding proteins can interfere with the hormonemeasurement by either ‘capturing’ the tracer mole-cule in a competitive assay or by preventing anti-body binding (Fisker & Orskov 1996). Dependingon assay type, the interference from binding pro-teins in the sample will lead to under or overesti-mation of the hormone concentration. A source ofinterference with no relation to the hormone ofinterest is the presence of heterophilic antibodies inthe patient’s sample. These antibodies can ‘link’ thecapture to the detection antibody directly, leadingto falsely increased assay results (Kricka 1999). Ithas been described that such antibodies can beinduced by pets housed by the patient (Park et al.2003), but in many cases the origin remains unclear.Methods have been developed to avoid such inter-ference, but the issue remains a problem (Emersonet al. 2003; Preissner et al. 2003). Finally, the matrixused to dissolve the calibrator rarely is identical tohuman serumain many cases, animal sera or bufferssupplemented with albumin are used. The beha-vior of antibodies sometimes can be influenced by

Page 56: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

40 chapter 4

matrix components, leading to differences in theassay results depending on the calibrator’s matrix.

Because all these factors can have profound effectson the hormone concentration measured, a carefulinterpretation of immunoassay results is required.Every single method must be evaluated in terms ofcross-reacting substances, and ideally the exactnature of the epitope recognized by the antibodiesinvolved is known. Obviously, ‘references ranges’for hormone concentrations are method dependent(Strasburger et al. 1996, 2001; Stenman et al. 1997;Quarmby et al. 1998; Wood 2001; Sharpe et al. 2002),and the simple reference to ‘general’ normative datafrom textbooks should be avoided. Finally, there isstill a need for the development of IRPs for manypeptide hormones. As demonstrated most recentlyfor hCG (Birken et al. 2003), the development of suitable reference reagents is a sophisticated task,but helps to eliminate some factors of uncertainty inimmunoassay measurements.

Peptide hormone measurements indoping tests

The Olympic Movement Anti-Doping Code as of January 1st, 2003 contains the statement that ‘the presence of an abnormal concentration of anendogenous hormone in class (E) or its diagnosticmarker(s) in the urine of a competitor constitutes an offence unless it has been proven to be due to a physiological or pathological condition’ (seewww.wada-ama.org/docs). Unfortunately, in thecase of peptide hormones the definition of an‘abnormal concentration’ is extremely difficult oreven impossible in many cases. Several peptide hormones are secreted by the human body in a pulsatil rather than a continuous manner, or thesecretion exhibits a circadian profile. Furthermore,their concentration is usually influenced not only by age and gender but also by environmental fac-tors (temperature, altitude), stress (psychological or physiological), sleep, nutrition state or training status. Many peptide hormones show an extremelyshort half-life time, leading to highly floatable concentrations in circulation. Therefore, a simplemeasurement of the hormone concentration in onlya few cases (like hCG in men) is sufficient to demon-strate the misuse of the hormone. In contrast to

some reports occurring in the laymen’s press, a high concentration of hGH, for example, by no means issufficient to prove that an athlete has been usingrecombinant hGH (Armanini et al. 2002).

Another problem with the detection of dopingwith peptide hormones is their recombinant origin.Derived from the expression of the protein encodedby the human gene sequence transferred to an invitro system, the artificially produced hormones are identical to the naturally occurring hormone intheir amino acid sequence and thus in their phy-sicochemical properties. Once a peptide hormonewas injected, for many years it was impossible to judge about the origin of a single hormonemolecule.

Detecting peptide hormone doping is further com-plicated by the fact thatacompared to the rathersimple, small and stable steroid hormone moleculesapeptide hormones are larger molecules exhibitinga very sensitive three-dimensional structure. Inmany cases, peptide hormones are rapidly degraded,metabolized and cleaved. The renal excretion in-volves complex processes, many of them being stillpoorly understood. Furthermore, the peptide hor-mone concentrations found in urine are often evenmuch lower than those in blood. Therefore, thematerial traditionally used in doping analyticsa

urineais of limited value for many peptide hor-mone tests. Blood sampling is required, bearing allthe ethical and legal problems discussed elsewhere(Birkeland & Hemmersbach 1999).

Finally, the problem to establish doping tests forpeptide hormones is related to the methodologicaldifference in the measurement of peptide versussteroid hormones outlined above. For several years,steroid hormones have been identified by gas chro-matography/mass spectrometry (GC/MS), andcutting edge experience together with the appro-priate equipment is present in the InternationalOlympic Committee (IOC) accredited laboratories.In many cases, GC/MS determination is referred toas a ‘reference technology’ or ‘gold standard’ for thequantitation of hormones. Unfortunately, this tech-nology has not been available for peptide hormoneanalysis for many years, and there still is no estab-lished procedure shown to be ready to use in dopingdetection (Bowers 1997; Hilderbrand et al. 2003). Allmethods currently available or at least expected to

Page 57: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

measurement of peptide hormones 41

be available in the near future rely on immunoas-say techniques, in the case of EPO complemented by an electrophoretic confirmation based on isoelectricfocusing (IEF).

Specific aspects in doping analytics

Chorinonic gonadotropin

Chorinonic gonadotropin (hCG) is an importantpregnancy-associated hormone that stimulatesendogenous steroid production. The abuse of hCGin male athletes has been described, aiming toenhance endogenous anabolic steroids withoutchanging the testosterone/epitestosterone ratio(Stenman et al. 1997). Normally, hCG concentrationsin non-pregnant women and especially in men are extremely low. Only in a few pathological situations such as testicular cancer are know to beaccompanied by elevated concentrations in men(Lottersberger et al. 2003). Therefore, the case ofhCG is a comparably simple situation in peptidehormone doping analytics: The presence of highhCG levels in a male athlete are highly suspiciousfor doping. However, even frequently used in clin-ical practice, the quantification of hCG remainsproblematic. As a member of the glycoprotein hor-mone family, hCG is comprised of two distinct subunits (α and β). The α subunit is shared with allother members of this hormone family, whereas theβ subunit is specific and responsible for receptorbinding and biological activity. The carboxyl termi-nal of the hCG molecule contains a highly glycosy-lated region. Several molecular isoforms of hCGhave been identified, and their heterogeneity con-tributes substantially to the large between-methoddifferences in existing hCG immunoassays (Cole &Kardana 1992; Cole 1997; Cole et al. 2001). The situa-tion is even worse in urine, where the spectrum ofisoforms and degradation products is more com-plicated than in serum (Birken et al. 1996; O’Connoret al. 1999).

In addition to the heterogeneity of the analyte in a sample, for many years the standard prepara-tions used to calibrate hCG assays contained sub-stantial amounts of contaminating variants of hCG,which reacted to a variable degree with the anti-bodies in different immunoassays. It has been only

recently that an international study group was ableto establish six different IRPs for the most importanthCG isoforms (Birken et al. 2003), and their intro-duction is expected to make assay results more comparable.

Most recently, a tandem mass spectrometric ana-lysis technique has been described for confirmationof positive hCG tests (Gam et al. 2003). Followingtryptic digestion, it was possible to identify amarker peptide (βT5) providing a specific finger-print for hCG. Combined with a specific immunoex-traction procedure, the proposed method currentlyis able to quantify hCG concentrations as low as 5 IU·mL–1 Whether this MS approach in the field ofpeptide hormone analysis will be reliable and practicable in doping analytics remains to demon-strated in large-scale studies.

Erythropoietin

The introduction of a doping test for recombinanthuman erythropoietin (rhEPO) at the 2000 SydneyOlympic games represents a major step forward inthe fight against doping (Kazlauskas et al. 2002). Forthe first time, a detection method for a peptide hor-mone able to discriminate between the recombinantand the endogenous form of the hormone has beenimplemented into the official doping test program.To the present, detection of doping with rhEPOrelies on two different methods. The first is the urine-based test as described by Lasne and de Ceaurriz(Lasne & de Ceaurriz 2000; Lasne et al. 2002), whichutilizes an IEF method in combination with a tech-nique called ‘double-blotting’ enabling the visual-ization of protein bands with a greatly reducedbackground staining (Lasne 2001, 2003). The ration-ale of the test is to detect the pattern of isoforms ofEPO in a sample, which is different between recom-binant and endogenous EPO (Lasne et al. 2002; De Frutos et al. 2003). The reason seems to be thatespecially glycosylation are sensitive to the cellularenvironment where a protein is produced. BecauserhEPO is produced mainly in Chinese hamsterovary cells, the pattern of glycosylation is differentfrom that in the human kidney (Sasaki et al. 1987;Rice et al. 1992). Meanwhile, this test procedure hasbeen implemented in several IOC accredited labor-atories and is used in doping analyses from many

Page 58: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

42 chapter 4

sports associations (Catlin et al. 2002; Breidbach et al.2003).

A major problem with this technique is that it isvery expensive and time-consuming. In addition,the likelihood of catching a cheating athlete is givenonly for the first 3–4 days after injection (Wide et al.1995; Souillard et al. 1996). The attempts to circum-vent these problems led to the development ofmethods based on detecting changes in hematologicparameters after injection of rhEPO. These changeshave been demonstrated to be more pronouncedthan can be expected from the normally observedvariability, and some of them are detectable for as long as 4 weeks after rhEPO administration(Parisotto et al. 2001; Gore et al. 2003). The so-called‘second generation blood test’ involves the measure-ment of EPO and of the soluble transferin receptor,both done by immunoassay techniques. Being mucheasier, cheaper and providing a much greater window of opportunity to detect rhEPO misuse, this test procedure unfortunately relies on bloodsamples and gives a comparably high rate of falsepositives when the cut off is set to a level where not too many abusers are missed. Thus, a currentapproach is to use the blood-based tests as a screen-ing test to identify those samples which are morelikely to contain rEPO, followed by the urine-basedIEF test as a confirmatory test applied only to suspi-cious samples. Gore et al. (2003) have clearly shownthat using the second-generation blood test forscreening can drastically reduce the costs per posit-ive sample as judged by the urine test. In addition, a lot of work has been done to establish normativedata and to identify factors potentially influencingthe test outcome (Sharpe et al. 2002; Ashenden et al.2003; Parisotto et al. 2003). However, it is importantto remember that the EPO molecules found in aurine sample can only be proven to be of recombin-ant origin by the IEF urinary approach visualizingthe typical glycosylation pattern.

Growth hormone and growth hormone-dependent hormones

The detection of doping with recombinant humangrowth hormone (rhGH) has been thought to beimpossible for many years. However, recently it has

been shown that at least two different approachesare able to discriminate whether or not an athletehas taken rhGH. The test methods developed arestill in the validation process and not yet imple-mented as an official doping test, but this isexpected to happen soon.

A major problem remains that in contrast to thesituation with EPO, none of the proposed methodsfor detecting hGH doping today can be applied inurine samples. This is primarily due to the extremelylow hGH concentrations found in urine making theanalysis not possible by currently available tech-niques. Furthermore, hGH secretion to urine is acomplex process which seems to be highly variableand is still poorly understood (Saugy et al. 1996).The IEF method used for EPO in urine samples isnot useful in the case of hGH, because hGH does not contain any glycosylation sites. According toour knowledge today, the hGH molecules of recom-binant origin are almost identical to the main frac-tion of hGH molecules secreted by the pituitarygland, and no distinct physico-chemical propertiesof rhGH have been described.

However, even no glycosylation sites are present;hGH in circulation consists of a mixture of molecu-lar isoforms (Baumann 1999). The investigation ofthese isoforms is not as advanced as in case of hCG,but during the last years it was possible to identifyat least some of the major components. In additionto the 22 kDa major isoform, consisting of 191 aminoacids, a shorter 20 kDa hGH isoform lacking aminoacids 32–46 is the second most abundant form ofhGH in circulation (Hashimoto et al. 1998; Tsushimaet al. 1999; Leung et al. 2002). There are other evenshorter isoforms described, but they are observedless constantly and are not yet fully analyzed. Someof them have been shown to be cleaved or degradedhGH molecules. The isoforms of hGH seem to existin monomeric, dimeric and multimeric complexesformed by either identical (homodimers) or differ-ent isoforms (heterodimers).

Many of the hGH effects in the body are mediatedthrough a factor called insulin-like growth factor I(IGF-I). Produced mainly in the liver, but evenlocally in cartilage, bone and many other tissues,IGF-I is secreted to the blood, where it is bound by specific binding proteins (Le Roith et al. 2001).

Page 59: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

measurement of peptide hormones 43

The most important ones are IGF binding protein-3(IGFBP-3) and the acid labile subunit (ALS), bothproduced under hGH control as well. At least forIGFBP-3 it has been shown that this protein exertseffects independently from IGF-I binding and there-fore can be a seen as a ‘peptide hormone’ itself.Together, IGF-I, IGFBP-3 and ALS form a 150 kDaternary complex, possessing an increased half-life incomparison to each molecule alone.

It was one strategy in the search for a suitable testmethod to detect hGH doping to evaluate whetheror not the increase in the pharmacodynamic end-points of hGH action, especially the increase in thecomponents of the ternary complex might exceedthe normally observed variability (Dall et al. 2000).Such a test would include the advantage that thehalf-life of the pharmacodynamic endpoints of hGHaction exceeds that of hGH, making possible alonger window of opportunity for detecting hGHabuse. An international consortium of researchersconducted a series of large-scale studies to invest-igate the behavior of such pharmacodynamic endpoints of hGH action in relation to various con-ditions like acute and chronic exercise, age, gender,ethnic background or injury (Wallace et al. 1999,2000; Longobardi et al. 2000; Ehrnborg et al. 2003).The main outcome of these studies was that re-peated administration of exogenous rhGH indeedinduces changes in pharmacodynamic endpoints,which can be discriminated from changes inducedby exercise or other stimulators. In more detail, theresearchers developed a statistical model includingmore than one of the pharmacodynamic endpointsand tailored this model specifically for each gender.However, a crucial point is that the immunoassaysused for determinations of the factors are suffi-ciently evaluated and that method specific referenceranges are known. Not all commercially availablemethods can fulfill these criteria, and therefore acareful selection of the immunoassays to be used ismandatory. Furthermore, because a highly sophist-icated statistical model is the backbone of this test method, the variability of the assays must beknown, and a continuous supply with an identicalbatch of antibodies must be ensured. This could pro-vide problems because many of the immunoassaysused until today rely on polyclonal rather than mono-

clonal antibodies. Polyclonal antisera, however, areinherently limited in their quantity andaonce abatch is finishedacannot be reproduced identically.Therefore, one can understand that the internationalantidoping agencies put the development of suit-able monoclonal antibodies onto the agenda. Oncethe methodological details have been clarified, thistest method could be very useful, comparably to theblood test already in use for EPO doping detection.

Another approach more directly relies on themeasurement of hGH: In contrast to the hGH isoform spectrum secreted by the pituitary gland,rhGH as produced in bacteria usually consist of 22 kDa hGH only. The recombinant production of20 kDa hGH has been described, but until today this preparation has been used only in a few clinicaltrials. Recombinant hGH used to treat GH deficientchildren, adolescents and adults is 22 kDa hGH, andthe same seems to apply for the hGH preparationsabused for doping purposes in sports. This ‘unifor-mity’ or ‘lack of heterogeneity’ in the recombinanthGH preparations in comparison to the many iso-forms secreted by the human pituitary gland andnormally present in circulation forms the basis forthe so-called ‘differential immunoassay approach’to detect doping with recombinant hGH (Wu, Z. et al. 1999): the injection of recombinant monomeric22 kDa hGH increases the relative abundance of thishGH isoform in circulation. This change in the iso-form composition is further increased by the reduc-tion of pituitary hGH secretion due to the negativefeedback mechanism observed under chronic rhGHtreatment (Wallace et al. 2001a). Screening of mono-clonal antibodies raised against different hGHpreparations led to the development of two differ-ent immunoassays for hGH. The capture antibodyof the first assay preferentially binds the 22 kDa iso-form of human GH, whereas the capture antibodyof the second assay preferentially binds ‘pituit-ary derived hGH’, consisting of multiple isoforms(Bidlingmaier et al. 2000). Measuring on serum sam-ple by both assays allows to calculate the relativeabundance of 22 kDa hGH versus the other isoforms(‘total hGH’), and thus identifying samples contain-ing an inappropriately high content of 22 kDa hGH.It could be demonstrated that the change in themolecular isoform composition is uniquely related

Page 60: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

44 chapter 4

to rhGH application, whereas the increase in cir-culating hGH after exercise is accompanied by anincrease in the non-22 kDa hGH isoforms as well(Wallace et al. 2001b). Meanwhile, the originalapproach has been dramatically improved in termsof sensitivity (Bidlingmaier et al. 2003). This wasmade possible by the identification of new mono-clonal antibodies. In addition, an independentconfirmatory set of assays was developed, based on new monoclonal capture antibodies, targetingindependent epitopes. The latter is a prerequisite for the acceptability of immunoassays in a doping testsetting: each assay has to be confirmed by anotherassay, targeting independent structures of the mole-cule of interest and thus providing further evidencefor the identity of the molecule.

Inherently, the differential immunoassay ap-proach is limited to the detection of rhGH abusea

cadaveric hGH preparations, derived from extrac-tion of pituitary glands, have an isoform patternwhich cannot be discriminated from that naturallyoccurring in the human body. Furthermore, due tothe very short half-life time of hGH in circulation(about 15 min), the window of opportunity for thedetection remains limited to 24–36 h. Obviously,even the development of more sensitive assays willnot overcome this limitation, because after dis-appearance of the recombinant substance and cessa-tion of the negative feedback, the normal pituitaryisoform secretion has been shown to start again. Onthe other hand the fact that rhGH has to be taken bydaily subcutaneous injections enhances the prob-ability to catch a cheating athlete in unannouncedout-of-competition tests.

The differential immunoassay approach alsomakes it necessary to rigorously validate theimmunoassays selected. Furthermore, because aratio is calculated, the within- and between-method

variability must be determined exactly with respectto the potential impact of this variability on the ratio result. A reduction of the variability could beachieved by using the same capture antibody forboth, the 22 kDa preferential and the total hGHassay: on one microtiterplate, the upper half is covered by the 22 kDa assay capture monoclonalantibody (mAb), whereas the lower half of the plateis covered by the total hGH capture mAb. Afteradding calibrators, controls and samples to eachhalf of the plate, the whole plate is covered by thesame detection mAb. This reduces significantly thevariability always included in distributing sampleson two different plates (Bidlingmaier et al. 2000).

Conclusions

The immunoassay techniques used for peptide hor-mone measurements represent very sensitive toolsto quantify the concentrations of the hormones invarious biological fluids specifically. Appropriatelyselected antibodies, which must be characterizedregarding their epitopes, affinities and possiblycross-reacting peptides, confer a high degree ofspecificity in hormone measurements, making thesemethods suitable even in the setting of doping testprocedures. Being much easier, cheaper and lesstime consuming than other methods for peptidehormone analysis, immunoassays have a highpotential, especially in large-scale screening. How-ever, for each assay used, method specific norm-ative data have to be established. Furthermore, potentially confounding factors must be identifiedand appropriate methods for their elimination mustbe developed. If these precautions are taken intoconsideration, the accuracy of immunoassay ana-lyses can be extremely high and very suitable for use in a forensic environment.

References

Annesley, T.M. (2003) Ion suppression inmass spectrometry. Clinical Chemistry49(7), 1041–1044.

Armanini, D., Faggian, D., Scaroni, C. & Plebani, M. (2002) Growth hormoneand insulin-like growth factor I in aSydney Olympic gold medallist.

British Journal of Sports Medicine 36(2),148–149.

Ashenden, M.J., Gore, C.J., Parisotto, R. et al. (2003) Effect of altitude on second-generation blood tests to detecterythropoietin abuse by athletes.Haematologica 88(9), 1053–1062.

Baumann, G. (1999) Growth hormoneheterogeneity in human pituitary andplasma. Hormone Research 51 (suppl. 1),2–6.

Baumann, G., Amburn, K. & Shaw, M.A.(1988) The circulating growth hormone(GH)-binding protein complex: a major

Page 61: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

measurement of peptide hormones 45

constituent of plasma GH in man.Endocrinology 122(3), 976–984.

Bidlingmaier, M., Wu, Z. & Strasburger,C.J. (2000) Test method: GH. Bailliere’sBest Practice and Research. ClinicalEndocrinology and Metabolism 14(1),99–109.

Bidlingmaier, M., Wu, Z. & Strasburger,C.J. (2003) Problems with GH doping in sports. Journal of EndocrinologicalInvestigation 26, 924–931.

Binz, P.A., Hochstrasser, D.F. & Appel,R.D. (2003) Mass spectrometry-basedproteomics: current status and potentialuse in clinical chemistry. ClinicalChemistry and Laboratory Medicine 41(12),1540–1551.

Birkeland, K.I. & Hemmersbach, P. (1999)The future of doping control in athletes.Issues related to blood sampling. SportsMedicine (Auckland, NZ) 28(1), 25–33.

Birken, S., Kovalevskaya, G. & O’Connor,J. (1996) Metabolism of hCG and hLH tomultiple urinary forms. Molecular andCellular Endocrinology 125(1–2), 121–131.

Birken, S., Berger, P., Bidart, J.M. et al.(2003) Preparation and characterizationof new WHO reference reagents forhuman chorionic gonadotropin andmetabolites. Clinical Chemistry 49(1),144–154.

Black, R.S. & Bowers, L.D. (2000) Massspectrometric characterization of the β-subunit of human chorionicgonadotropin. Methods in MolecularBiology 146, 337–354.

Boguszewski, C.L. (2003) Molecularheterogeneity of human GH: from basicresearch to clinical implications. Journalof Endocrinological Investigation 26(3),274–288.

Bowers, L.D. (1997) Analytical advances in detection of performance-enhancingcompounds. Clinical Chemistry 43(7),1299–1304.

Breidbach, A., Catlin, D.H., Green, G.A. et al. (2003) Detection of recombinanthuman erythropoietin in urine byisoelectric focusing. Clinical Chemistry49(6 Pt. 1), 901–907.

Catlin, D.H., Breidbach, A. Elliott, S. &Glaspy, J. (2002) Comparison of theisoelectric focusing patterns ofdarbepoetin alfa, recombinant humanerythropoietin, and endogenouserythropoietin from human urine.Clinical Chemistry 48(11), 2057–2059.

Cole, L.A. (1997) Immunoassay of humanchorionic gonadotropin, its freesubunits, and metabolites. ClinicalChemistry 43(12), 2233–2243.

Cole, L.A. & Kardana, A. (1992) Discordant results in human chorionicgonadotropin assays. Clinical Chemistry38(2), 263–270.

Cole, L.A., Shahabi, S., Butler, S.A. et al.(2001) Utility of commonly usedcommercial human chorionicgonadotropin immunoassays in thediagnosis and management oftrophoblastic diseases. Clinical Chemistry47(2), 308–315.

Dall, R., Longobardi, S., Ehrnborg, C. et al. (2000) The effect of 4 weeks ofsupraphysiological growth hormoneadministration on the insulin-likegrowth factor axis in women and men.GH-2000 Study Group. Journal of ClinicalEndocrinology and Metabolism 85(11),4193–4200.

De Frutos, M., Cifuentes, A. & Diez-Masa,J.C. (2003) Differences in capillaryelectrophoresis profiles of urinary andrecombinant erythropoietin.Electrophoresis 24(4), 678–680.

Ehrnborg, C., Lange, K.H., Dall, R. et al.(2003) The growth hormone/insulin-likegrowth factor-I axis hormones and bonemarkers in elite athletes in response to amaximum exercise test. Journal of ClinicalEndocrinology and Metabolism 88(1),394–401.

Ekins, R. (1991) Immunoassaystandardization. Scandinavian Journal of Clinical and Laboratory Investigation.Supplementum 205, 33–46.

Emerson, J.F., Ngo, G. & Emerson, S.S.(2003) Screening for interference inimmunoassays. Clinical Chemistry 49(7),1163–1169.

Fisker, S. & Orskov, H. (1996) Factorsmodifying growth hormone estimates in immunoassays. Hormone Research46(4–5), 183–187.

Gam, L.H., Tham, S.Y. & Latiff, A. (2003)Immunoaffinity extraction and tandemmass spectrometric analysis of humanchorionic gonadotropin in dopinganalysis. Journal of Chromatography. B,Analytical Technologies in the Biomedicaland Life Sciences 792(2), 187–196.

Gore, C.J., Parisotto, R. Ashenden, M.J. et al. (2003) Second-generation bloodtests to detect erythropoietin abuse byathletes. Haematologica 88(3), 333–344.

Hashimoto, Y., Ikeda, I., Ikeda, M. et al.(1998) Construction of a specific andsensitive sandwich enzymeimmunoassay for 20 kDa human growthhormone. Journal of ImmunologicalMethods 221(1–2), 77–85.

Hilderbrand, R.L., Wanninger, R. &Bowers, L.D. (2003) An update on

regulatory issues in antidopingprograms in sport. Current SportsMedicine Reports 2(4), 226–232.

Kast, J., Parker, C.E., van der Drift, K. et al. (2003) Matrix-assisted laserdesorption/ionization directed nano-electrospray ionization tandem massspectrometric analysis for proteinidentification. Rapid Communications inMass Spectrometry 17(16), 1825–1834.

Kazlauskas, R., Howe, C. & Trout, G.(2002) Strategies for rhEPO detection insport. Clinical Journal of Sport Medicine12(4), 229–235.

Kricka, L.J. (1999) Human anti-animalantibody interferences in immunologicalassays. Clinical Chemistry 45(7), 942–956.

Lasne, F. (2001) Double-blotting: a solution to the problem of non-specificbinding of secondary antibodies inimmunoblotting procedures. Journal of Immunological Methods 253(1–2),125–131.

Lasne, F. (2003) Double-blotting: a solution to the problem of nonspecificbinding of secondary antibodies inimmunoblotting procedures. Journal of Immunological Methods 276(1–2),223–226.

Lasne, F. & de Ceaurriz, J. (2000)Recombinant erythropoietin in urine.Nature 405(6787), 635.

Lasne, F., Martin, L., Crepin, N. & deCeaurriz, J. (2002) Detection ofisoelectric profiles of erythropoietin inurine: differentiation of natural andadministered recombinant hormones.Analytical Biochemistry 311(2), 119–126.

Le Roith, D., Bondy, C., Yakar, S., Liu, J.L.& Butler, A. (2001) The somatomedinhypothesis: 2001. Endocrine Reviews22(1), 53–74.

Leung, K.C., Howe, C., Gui, L.Y. et al. (2002) Physiological andpharmacological regulation of 20-kDagrowth hormone. American Journal ofPhysiology. Endocrinology and Metabolism283(4), E836–843.

Liu, C. & Bowers, L.D. (1997) Massspectrometric characterization of nickedfragments of the β-subunit of humanchorionic gonadotropin. ClinicalChemistry 43(7), 1172–1181.

Longobardi, S., Keay, N., Ehrnborg, C. et al.(2000) Growth hormone (GH) effects onbone and collagen turnover in healthyadults and its potential as a marker ofGH abuse in sports: a double blind,placebo-controlled study. The GH-2000Study Group. Journal of ClinicalEndocrinology and Metabolism 85(4),1505–1512.

Page 62: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

46 chapter 4

Lottersberger, C., Hoermann, R., Mann, K.,Schwarz, S. & Berger, P. (2003) Tumor-and pregnancy-derived isoforms ofhuman chorionic gonadotropin:biological and diagnostic relevance.Hormone Research 59(3), 125–134.

Nagy, A.M., Meuris, S. & Robyn, C. (1994)Origin and significance of theheterogeneity of protein hormones.Nuclear Medicine and Biology 21(3),317–330.

O’Connor, J.F., Kovalevskaya, G. & Birken,S. (1999) Interference of luteinizinghormone β-core fragment in urinarygonadotropin assays. Clinical Chemistry45(12), 2290–2292.

Parisotto, R., Wu, M., Ashenden, M.J. et al.(2001) Detection of recombinant humanerythropoietin abuse in athletes utilizingmarkers of altered erythropoiesis.Haematologica 86(2), 128–137.

Parisotto, R., Ashenden, M.J., Gore, C.J. et al. (2003) The effect of commonhematologic abnormalities on the abilityof blood models to detect erythropoietinabuse by athletes. Haematologica 88(8),931–940.

Park, A., Edwards, M., Donaldson, M.,Ghatei, M. & Meeran, K. (2003) Lesson of the week: interfering antibodiesaffecting immunoassays in woman withpet rabbits. BMJ (Clinical Research Ed.)326(7388), 541–542.

Preissner, C.M., O’Kane, D.J., Singh, R.J.,Morris, J.C. & Grebe, S.K.G. (2003)Phantoms in the assay tube: heterophileantibody interferences in serumthyroglobulin assays. Journal of ClinicalEndocrinology and Metabolism 88(7),3069–3074.

Quarmby, V., Quan, C., Ling, V., Compton,P. & Canova-Davis, E. (1998) How muchinsulin-like growth factor I (IGF-I)circulates? Impact of standardization onIGF-I assay accuracy. Journal of ClinicalEndocrinology and Metabolism 83(4),1211–1216.

Ranke, M.B., Feldt-Rasmussen, U., Bang, P.et al. (2001) How should insulin-likegrowth factor I be measured? Aconsensus statement. Hormone Research55 (suppl. 2), 106–109.

Rice, K.G., Takahashi, N., Namiki, Y. et al.(1992) Quantitative mapping of the N-linked sialyloligosaccharides ofrecombinant erythropoietin,

combination of direct high-performanceanion-exchange chromatography and 2-aminopyridine derivatization. AnalyticalBiochemistry 206(2), 278–287.

Roger, M. & Lalhou, N. (1994)Heterogeneity of plasma gonadotropins.Consequences on immunologicalproperties of LH. Nuclear Medicine andBiology 21(3), 349–357.

Sasaki, H., Bothner, B., Dell, A. & Fukuda,M. (1987) Carbohydrate structure oferythropoietin expressed in Chinesehamster ovary cells by a humanerythropoietin cDNA. Journal ofBiological Chemistry 262(25), 12 059–12 076.

Saugy, M., Cardis, C., Schweizer, C.,Veuthey, J.L. & Rivier, L. (1996)Detection of human growth hormonedoping in urine: out of competition testsare necessary. Journal of Chromatography.B, Biomedical Applications 687(1),201–211.

Sharpe, K., Hopkins, W., Emslie, K.R. et al.(2002) Development of reference rangesin elite athletes for markers of alterederythropoiesis. Haematologica 87(12),1248–1257.

Souillard, A., Audran, M., Bressolle, F. et al. (1996) Pharmacokinetics andpharmacodynamics of recombinanthuman erythropoietin in athletes. Bloodsampling and doping control. BritishJournal of Clinical Pharmacology 42(3),355–364.

Stenman, U.H., Unkila-Kallio, L.,Korhonen, J. & Alfthan, H. (1997)Immunoprocedures for detectinghuman chorionic gonadotropin: clinicalaspects and doping control. ClinicalChemistry 43(7), 1293–1298.

Strasburger, C.J., Wu, Z., Pflaum, C.D. & Dressendorfer, R.A. (1996)Immunofunctional assay of humangrowth hormone (hGH) in serum: apossible consensus for quantitative hGH measurement. Journal of ClinicalEndocrinology and Metabolism 81(7),2613–2620.

Strasburger, C.J., Bidlingmaier, M., Wu, Z.& Morrison, K.M. (2001) Normal valuesof insulin-like growth factor I and theirclinical utility in adults. HormoneResearch 55 (suppl. 2), 100–105.

Tsushima, T., Katoh, Y., Miyachi, Y. et al.(1999) Serum concentration of 20 K

human growth hormone (20 K hGH)measured by a specific enzyme-linkedimmunosorbent assay. Study Group of 20 K hGH. Journal of ClinicalEndocrinology and Metabolism 84(1),317–322.

Wallace, J.D., Cuneo, R.C., Baxter, R. et al. (1999) Responses of the growthhormone (GH) and insulin-like growth factor axis to exercise, GHadministration, and GH withdrawal intrained adult males: a potential test forGH abuse in sport. Journal of ClinicalEndocrinology and Metabolism 84(10),3591–3601.

Wallace, J.D., Cuneo, R.C., Lundberg, P.A.et al. (2000). Responses of markers ofbone and collagen turnover to exercise,growth hormone (GH) administration,and GH withdrawal in trained adultmales. Journal of Clinical Endocrinologyand Metabolism 85(1), 124–133.

Wallace, J.D., Cuneo, R.C., Bidlingmaier,M. (2001a) Changes in non-22-kilodalton (kDa) isoforms of growthhormone (GH) after administration of22-kDa recombinant human GH intrained adult males. Journal of ClinicalEndocrinology and Metabolism 86(4),1731–1737.

Wallace, J.D., Cuneo, R.C., Bidlingmaier,M. (2001b) The response of molecularisoforms of growth hormone to acuteexercise in trained adult males. Journal of Clinical Endocrinology and Metabolism86(1), 200–206.

Wide, L., Bengtsson, C., Berglund, B. &Ekblom, B. (1995) Detection in blood andurine of recombinant erythropoietinadministered to healthy men. Medicineand Science in Sports and Exercise 27(11),1569–1576.

Wood, P. (2001) Growth hormone: itsmeasurement and the need for assayharmonization. Annals of ClinicalBiochemistry 38(Pt. 5), 471–482.

Wu, S.L., Amato, H., Biringer, R. et al.(2002) Targeted proteomics of low-levelproteins in human plasma by LC/MSn:using human growth hormone as amodel system. Journal of ProteomeResearch 1(5), 459–465.

Wu, Z., Bidlingmaier, M., Dall, R. &Strasburger, C.J. (1999) Detection ofdoping with human growth hormone.Lancet 353(9156), 895.

Page 63: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

47

Historical aspects of doping controlanalysis of low molecular weight drugs

Ever since sport competition has been known, thewish to artificially enhance power, skill and strengthwas also present. Philostratos and Galen alreadyreported attempts of athletes of the ancient OlympicGames to improve performance and endurance bymeans of concoctions of mushrooms and plant seedsas well as special diets, for example with bovine test-icles (Burstin 1963; Prokop 1970, 2002). Moreover,the athletes’ animals were targets of manipulationas described for the ancient Romans, who fed theirhorses hydromel, a mixture of honey and water, inorder to increase their strength (Morgan 1957;Ariëns 1965). Major reasons for the phenomenon ofathletes trying to artificially obtain the cutting edgeadvantage in competition are supposed to be fame,glory and honor as well as financial aspects, whichbecame even more important with the introductionof professional sports and horse and greyhound racing. At the beginning of the 20th century, first scientific procedures for the determination of doping agents in horse saliva were developed byRussian and Austrian scientists, and in 1910–1911about 220 samples were investigated regardingadministration of alkaloids. Between 1938 and 1954,principal procedures for the detection of stimulantssuch as amphetamines were presented, whichdemonstrated limited sensitivity and susceptibilityto interferences by biological matrices (Richter 1938; Keller & Ellenbogen 1952; Axelrod 1954), butin 1956, a commonly accepted assay based on liquidextraction, paper chromatography and visualiza-tion was introduced (Vidic 1956). The fatality of

doping became obvious in 1886, when the cyclistLinton died during a race Paris–Bordeaux caused by an overdose of caffeine (Prokop 2002). In the fol-lowing 70 years, numerous cases of doping-relateddeaths were documented, primarily involvingcyclists, such as the death of a cyclist caused byamphetamine poisoning (1949); the hospitalizationof a cyclist for confusional toxical state from excess-ive use of amphetamines (1956); a cyclist in state of shock from excessive use of sympathomimetics(1958); and hospitalization of a cyclist for intoxica-tion with amphetamines and analeptics (1962). Allthese cases were uncovered in Italy (Venerando1963). With the formation of doping commissions inFrance (1959), Austria (1962) and Italy (1963) followedby the Medical Sub-Commission of the InternationalOlympic Committee (IOC) in 1967, a comprehens-ive fight against doping was started. First officialdoping controls were conducted during the OlympicGames in Grenoble (1968) based on a first screeningassay for a selection of stimulants (Beckett et al.1967), and the list of prohibited substances andmethods of doping has been modified and extendedmany-fold during the last decades. A huge varietyof methods have been established in order to deter-mine doping-relevant compounds, their metabol-ites and their influence on endogenous parameters.

Classification of drugs

Prohibited compounds

Owing to the wide variety of banned compounds,there is no list mentioning all restricted substancesby name. Besides examples for each category, the

Chapter 5

Analysis of Low Molecular Weight Substancesin Doping Control

MARIO THEVIS AND WILHELM SCHÄNZER

Page 64: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

48 chapter 5

extension ‘. . . and related substances . . .’ is added,resulting in prohibition of all compounds with correlated pharmacological or physicochemicalproperties. In Table 5.1, the classification of drugsaccording to the IOC and World Anti-DopingAgency (WADA) (International Olympic Com-mittee 2003) is shown. For several remedies andsubstances such as beta-blockers, corticosteroids,local anesthetics, marihuana and alcohol, restrictionis limited to selected sport sections or their medicalindication.

stimulants

Stimulants are most likely the oldest doping sub-stances known to human beings. They include com-

pounds such as amphetamines, ephedrines, caffeineand also cocaine (Scheme 5.1). The latter wasalready used by Mexican Incas in the 16th centurywhen they utilized coca leaves to cope with dis-tances of more than 1000 miles (1609 km) betweenCuzco and Quito within 5 days (www.g-o.de 2003),and the very first identified doping substancesfound in athletes’ urine samples in the 20th centurywere related to amphetamines. Due to the presenceof ephedrines in a series of cold medicines, and fur-thermore the caffeine content of several cold and hot beverages, urinary concentration limits for theseselected compounds were established, requiringquantitation of these drugs in urine specimens,whereas new regulations valid from January 2004do not prohibit caffeine anymore.

narcotics

The class of narcotics includes compounds referredto as opioid-like analgesics such as morphine(Scheme 5.2) and related substances with a fewexemptions, for example ethylmorphine and co-deine, owing to their weak analgesic effect. Othernon-opioid-like drugs, for instance acetylsalicylicacid or diclofenac, are allowed.

Table 5.1 Classification of prohibited compoundsaccording to the International Olympic Committee (IOC)and World Anti-Doping Agency (WADA).

I. Prohibited classes of substancesA. StimulantsB. NarcoticsC. Anabolic agentsD. DiureticsE. Peptide hormones, mimetics and analogsF. Agents with anti-estrogenic activityG. Masking agents

II. Prohibited methodsA. Enhancement of oxygen transferB. Pharmacological, chemical and physical manipulationC. Gene doping

III. Classes of prohibited substances in certain sportsA. AlcoholB. CannabinoidsC. Local anestheticsD. GlucocorticosteroidsE. Beta-blockers

Scheme 5.1 Structure formulae of amphetamine (1), ephedrine (2), cocaine (3) and caffeine (4).

Scheme 5.2 Chemical structure of morphine, arepresentative of the class of opioid-like narcotics.

Page 65: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 49

anabolic agents

While stimulants and narcotics are of interest prim-arily in competition, anabolic agents are effectiveduring periods of exercise. Increasing muscle growthand strength gaining, the drugs belonging to thiscategory were added to the list of prohibited com-pounds of the IOC in 1976, and, since 1993, not onlyare steroids related to testosterone (Fig. 5.1) includedin the class of anabolic agents but also drugs such asclenbuterol, as β2-agonists cause anabolic effects ifadministered in doses significantly higher than fortherapeutic use (Reeds et al. 1988; Wagner 1989;Stallion et al. 1991; Höher & Troidl 1995).

diuretics

Diuretics are considered as doping agents based ontwo facts: (i) athletes competing in sports categor-ized by weight can decrease their body weight by artificially induced diuresis, i.e. increased urineflow; (ii) the increased loss of water dilutes the urineresulting in decreased concentration of excretedcompounds. Regarding those drugs that are bannedwith respect to cutoff limits, diuretics can mask the abuse of prohibited substances. The class ofdiuretics in particular demonstrates the possiblephysicochemical heterogeneity of one category ofdrugs. Diuretic agents can be divided into at leastsix groups owing to their structures, place andmechanism of effect. There are so-called thiazides(e.g. hydrochlorothiazide), sulfamoylbenzoic acidderivatives (e.g. furosemide), osmotic diuretics (e.g.mannitol), carbonic anhydrase inhibitors (e.g. ace-

tazolamide), phenoxyacetic acid derivatives (e.g.ethacrynic acid), and potassium-retaining diuretics(e.g. spironolactone), some of which are shown inScheme 5.3 (Wilhelm & deStevens 1976; Lang &Hropot 1995; Möhrke & Ullrich 1995).

beta-blockers

Beta-receptor blocking agents (also referred to asbeta-blockers) are banned for sports such as shoot-ing or ski jumping owing to their sedative effects. Ingeneral, beta-blockers consist either of a phenyl ringstructure bearing an oxypropanolamine side chainthat terminates in an isopropyl or tert. Butyl residue(e.g. propranolol, levobunolol), or a phenyleth-anolamine nucleus comprising substitutes such asnitrite functions (e.g. nifenalol). Modern β-receptorblocking agents partially diverge from this principalstructure, for instance the remedy nebivolol. Since1988 the class of beta-blockers belongs to the IOC listof prohibited compounds, and some examples arepresented in Scheme 5.4.

As shown in Table 5.1, additional classes of prohib-ited compounds, such as peptide hormones, belongto the list of banned substances, but in this overviewwe will focus only on a selection of low molecularweight drugs.

Analytical techniques

The first commonly accepted screening andconfirmation methods for doping control analysisincluded sample preparations based on liquid–liquid

1-methylation

1,2-oxidation

19-desmethylation

17-methylation

7-methylation

OH

O

2

3 45

67

8

14

18

18

1617

15

1112

9

19

1012-methylation

4,5-reduction

4-chlorination

Testosterone

Fig. 5.1 Chemical structure oftestosterone and modificationsresulting in various syntheticanabolic steroids.

Page 66: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

50 chapter 5

extraction of urine, concentration of the extracts and separation of analytes by means of gas–liquidchromatography (GLC) and thin-layer chromato-graphy (TLC). A flame ionization detector (FID) wasinterfaced to GLC indicating the presence of stimu-lants of the amphetamine type by signals at com-parable retention times as observed with referencecompounds. Thin-layer chromatography was usedfor the identification of strychnine and also ring-hydroxylated stimulants such as p-hydroxyam-phetamine and phenylephrine (Beckett et al. 1967).In case of ‘positive’ test results, additional informa-tion on the analytes was obtained by derivatization,micro infrared spectroscopy and also mass spectro-metry (MS). With improvements and innovations in analytical techniques, commercially available

instruments and sophisticated sample preparationprocedures, numerous assays have been developedenabling a comprehensive analysis of elite athlete’surine samples for doping control purposes. Depend-ing on the nature of compounds to be analyzed, different analytical tools have been employed, theprincipal techniques of some of which will bedescribed in the following.

In general, chromatographic separation of extractsobtained from biological material such as blood,urine or hair, is more or less obligatorily precedingthe analytical detection. Thus, more detailed infor-mation on analytes is provided; for example, byretention times that support for instance the distinc-tion between stereoisomers, or by separating lowlyconcentrated from highly abundant compounds.

Scheme 5.3 Structure formulae of selected diuretics: (1)hydrochlorothiazide, (2) furosemide,(3) mannitol, (4) acetazolamide, (5) ethacrynic acid and (6)spironolactone.

Scheme 5.4 Structure formulae of selected beta-blockers: (1)propranolol, (2) levobunolol, (3)nifenalol and (4) nebivolol.

Page 67: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 51

Modern chromatographic systems are based eitheron capillary gas chromatography or high perform-ance liquid chromatography, both of which havebecome extremely sophisticated and elaborated sciences.

Gas chromatography

Nowadays, capillary columns are the first choice for gas chromatography (GC) in doping controlanalysis. Their excellent performance in terms ofcompound separation, robustness, and the enorm-ous variety of different stationary phases providesnumerous options for method development andpossibilities to cope with tasks such as compre-hensive screening assays or compound-specific analyses. The principal composition of a moderncapillary GC column is presented as cross section inFig. 5.2, and the basic scaffold is represented bythree distinct parts: (i) outer protective coating; (ii)fused silica tubing; and (iii) stationary phase.

Fused silica tubing. Fused silica is a synthetic, quartz-like glass of very high purity regarding metal oxidecontaminants. In general, fused silica bears a veryactive surface owing to its silanol functions, whichcan interact with polar groups of analytes, such ashydroxyl, carboxyl and thiol residues as well as prim-ary and secondary amines, causing peak tailing anda decreased peak intensity. Hence, a deactivation ofsilanol groups by appropriate chemical treatment is

performed, for instance by trimethylsilylation, andthe derivatized fused silica creates a suitable surfacefor being coated by the stationary phase.

Stationary phase. The stationary phase is the mostcrucial component in terms of retention, separa-tion and peak shape of analytes in GC. This par-ticular part of capillary columns consists of eitherpolysiloxanes/silicones, polyethylene glycols (PEGs)or so-called porous layer phases. The predomin-antly used stationary phases are the substitutedpolysiloxanes owing to their robustness and super-ior lifetime. In Fig. 5.3, the general chemical struc-ture of polysiloxane-based stationary phases ispresented, determined by the alternating silicon-oxygen backbone and two substitutes at each siliconatom. The structure and amount of the substitutescharacterizes each stationary phase, and primarilyfour groups are utilized, mixed in different ratios: (i) methyl-; (ii) phenyl-; (iii) cyanopropyl-; and (iv)trifluoropropyl residues. With the choice of sub-stitutes and their relative presence in the stationaryphase, the polarity of the GC column is defined aslisted in Fig. 5.4 for 14 common mixtures. Anotherstationary phase employed in GC is PEG that ischaracterized mainly by the molecular weight or thechain length of the polymer. A remarkable differ-ence of this type of material to polysiloxane phasesis the possibility to designate a certain pH range, i.e.acidic or alkaline columns are available demon-strating an improved capability to separate acidic or alkaline compounds, respectively. A main disad-vantage is the high sensitivity to oxygen, in particu-lar at elevated temperatures, resulting in destructionof the stationary phase and a correlated short life-time. Porous layer stationary phases consist of smallporous particles (e.g. polymers, aluminium oxide,molecular sieve) that are bound to the fused silicatubing by chemical linkers. Owing to their highaffinity of gas adsorption, these phases are utilizedprimarily for the chromatography of very volatilecompounds or gases, which usually demonstratepoor retention on stationary phases such as poly-siloxane or PEG, and thus require temperaturesbelow 35°C. With so-called PLOT columns (porouslayer open tubular), gas–solid adsorption is the primary separation mechanism enabling efficient

Fused silica tubingProtective coating

Stationary phase

Fig. 5.2 Principal composition of capillary gaschromatography columns.

Page 68: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

52 chapter 5

retention of very volatile substances, but the stronginteraction between stationary phase and analyteexcludes their use for less volatile substrates.

Outer coating. As fused silica tubing is fairly fra-gile, it requires a protective outer coating. For thatpurpose, polyimide is commonly employed andprovides two advantages: first, polyimide-coatedcolumns are robust, stable and do not require delic-ate handling, and second, it covers and fills flaws of the fused silica tubing, preventing the growth ofdefects.

After considering the composition of capillary GC column, two additional parameters impact thechromatographic performance remarkably: columndimensions, i.e. length, diameter and film thickness,and carrier gases.

While length and diameter mainly affect peak

resolution, film thickness primarily influences thecapacity of columns and the retention of analytes.The length of a column is directly proportional tothe number of theoretical plates, and as the resolu-tion is proportional to the square root of the numberof theoretical plates, resolution is also proportionalto the square root of column length. In other words,doubling the column length (and thus the numberof theoretical plates) does not improve resolution by100%, but in practise only by about 25–35%, and thereduction of column length by 50% causes a loss ofresolution of approximately 15–25%. In addition,the number of theoretical plates is inversely propor-tional to the column diameter. Hence, decreasingcolumn diameter increases efficiency of GC. Takingagain into account that resolution is proportional tothe square root of the number of theoretical plates,halving of the inner diameter also improves resolu-tion by 25–35%; for instance, by exchanging a col-umn with an inner diameter of 0.25 mm by 0.11 mm.The capacity of capillary columns is directly de-pending on the film thickness of the stationaryphase. While phases of 0.1–0.2 µm are appropriatefor 20–50 ng of analyte, 0.5 µm are necessary foramounts greater than 125 ng. Thus, film thicknessmust be adjusted to the expected load of com-pounds injected onto the GC column.

Also, carrier gas impacts the performance of GC,as commonly described by the Van Deemter curve.Frequently employed carrier gases are helium and

Si

CH3 CH3 CH3

CH3CH3 CH3

O Si O

Si

CH3

(CH2)3CN

CH3

CH3

O

Methyl-cyanopropyl Methyl-trifluoropropyl

Si O

Si O

Methyl-phenylMethyl

Si O

n

n m

Si

CH3

(CH2)2CF3

CH3

CH3

O Si O

n m

n m

Increasin

g p

olarity

100% methyl5% phenyl20% phenyl35% phenyl50% phenyl40–50% trifluoropropyl polyethylene glycol50% cyanopropyl80% cyanopropyl100% cyanopropyl

Fig. 5.3 General chemical structureof polysiloxane-based stationaryphases with selected substitutes(methyl-, phenyl-, cyanopropyl- and trifluoropropyl residues).

Fig. 5.4 Stationary phase polarities depending on thechoice of substitutes and their relative amounts.

Page 69: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 53

hydrogen, the latter one provides a superior optimal linear velocity resulting in an improvedperformance regarding resolution. The disadvant-age of hydrogen is definitely its highly flammablecharacter.

Liquid chromatography

Comparable to GC, liquid chromatographic access-ories provide a wide variety of dimensions, pack-ing materials and, as compounds are carriedthrough the analytical columns by liquids, a seriesof organic solvents and buffers. In addition, severalprinciples of liquid chromatography (LC) (normalphase, reversed phase, ion exchange, size exclusion)can be applied, depending on the nature of targetanalytes. In this brief overview, we will focus onlyon reversed phase chromatography (RPC) which isthe most frequently used system in doping controlanalysis.

Cartridges. In LC in particular, numerous diame-ters of column cartridges are available. With theimprovements in packing materials (i.e. stationaryphases, see below), the most commonly employeddimensions for small molecule analysis in dopingcontrol laboratories have been reduced to columnlengths of 30–120 mm and inner diameters of 1.0–4.6 mm compared to earlier days of high perform-ance liquid chromatography (HPLC) where longerand broader columns were utilized. The shorteninghas become an option with the highly selective andspecific mass analyzers, which enable separation ofcompounds by means of their mass spectra, andthus the need for optimal chromatographic distinc-tion between analytes has become minor important.In addition, performance of LC columns has beenincredibly ameliorated, facilitating peak separationin reduced time frames.

Stationary phase. As mentioned in the Gas chro-matography section above, the stationary phase isthe utmost crucial point in LC as well. Here, differ-ent parameters have to be considered such as par-ticle size (3–50 µm), carrier (spherical or irregularsilica), chemically bonded phases (e.g. C4, C8, C18),and pore size (50–4000 Å).

For characterization of column performance, thenumber of plates n (or the plate height h) is fre-quently used as a measure. The Van Deemter equa-tion in its simplest form describes the reciprocalrelation of particle size to plate height, which meansthat a decrease in particle size entails an improve-ment in chromatography performance, namely re-solution (Yamashita & Fenn 1984a; Engelhardt et al.1985). Hence, there is a bias to reduce particle sizeswith the use of very short columns in high speedchromatography in order to maintain required peakresolutions.

The most commonly employed carrier materialfor the preparation of stationary phases in RPC issilica. Commercially available silicas differ in phys-ical properties such as specific surface area, averagepore diameter, specific pore volume and shape.Assuming open cylindrical pores, the three firstmentioned variables are related by the equation

φ = 103 × 4Vp/Osp

with φ = average pore diameter (nm), Vp = specificpore volume (mL·g–1) and Osp = specific surface area (m2·g–1) (Scott 1982). Average surface areas areapproximately 300 m2·g–1 and pore diameters 10 nm,giving rise to an approximate specific pore volumeof 1 mL·g–1. Nowadays, almost exclusively spher-ical silica is utilized as carrier for reversed phase LC stationary phases, owing to the higher density com-pared to silica prepared by polymerization of silicicacid yielding irregular particle shapes. On the sur-face, the silica carrier bears hydroxyl groups (silanolfunctions), which are utilized to chemically bindphases and thus define the principal nature of theHPLC column. An enormous variety of phases hasbeen introduced to the market in order to providesuitable material for each chromatographic chal-lenge, for instance the frequently used phenyl-, C18-,C8- and C4-phases, or more polar material such ascyano-, diol- or amino-phases as demonstrated inFig. 5.5. In addition, even more sophisticated sys-tems enabling the separation of chiral moleculeshave been developed, and the most recent improve-ment in high-throughput analysis was achieved byso-called monolithic columns consisting of a singlepiece of an organic polymer or silica with flow-through pores prepared by direct polymerization of

Page 70: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

54 chapter 5

appropriate monomers. These materials providebetter stability and, more importantly, higher per-formance than conventional columns packed withparticles.

Mobile phases. In case of RPC, the stationary phase is the non-polar and the mobile phase the polarcomponent. Conventional RPC employs buffer sys-tems consisting of, for example, KH2PO4/H3PO4or NaH2PO4 in combination with organic solventssuch as methanol or acetonitrile, which are verysuitable for analyzers such as ultraviolet (UV) -detectors. But the coupling of HPLC to MS requiresdifferent considerations regarding the eluents asthere are restrictions on pH, solvent choice, solventadditives and flow rates for LC in order to accom-plish optimal mass spectrometric results (Wheeler1955). In general, atmospheric pressure ionization(e.g. electrospray ionization, ESI) demands volatilesolvent additives to prevent ion source contamina-tions; hence commonly used phosphate, borate or

sulfate additives are not recommended but, forinstance, ammonium acetate, ammonium formateor tetraethy-lammonium hydroxide (TEAH) are.Moreover, ESI is not compatible with ingredientsfavoring the generation of strong ion pairs resultingin neutralization of ions after desorption. Control ofpH is of paramount importance in particular withESI, because of its enhancing effect on analyte ion-ization. Compounds of basic character should beanalyzed under acidic conditions utilizing additivessuch as acetic acid or formic acid in the 0.1–1.0% range, while components of acidic nature (e.g.carboxylic acids) are preferably negatively ionizedunder alkaline conditions; for example, by means of addition of ammonium hydroxide. Commonlyemployed organic solvents are comparable to con-ventional HPLC without interfacing to MS, i.e.methanol, ethanol and acetonitrile.

Detectors

The detection and identification of chromatographic-ally separated compounds is of paramount import-ance in doping control analysis. Many differenttypes of detectors have been developed such as FID,nitrogen phosphorus detector (NPD), UV-detectorand mass selective detectors such as quadru-pole, time-of-flight, ion trap, Fourier-Transform ioncyclotron resonance (FT-ICR), triple-stage quad-rupole and sector mass analyzers. In addition, hybridinstruments composed by two or more of theseunits are commercially available, and numerousapplications enabling the determination of adminis-tration of prohibited compounds are based on acombination of analyte-specific sample preparation,chromatography and sensitive as well as selectivedetectors.

flame ionization detector

One of the oldest and universal detectors inter-faced to GC is the flame ionization detector (FID)(Fig. 5.6). Here, the gas mixture eluting from the GCcolumn is aerated, fortified with hydrogen andignited electrically. The hydrogen–air flame itselfcreates only very few ions, but most organic com-pounds generate an increased number of ions and

Fig. 5.5 Commonly used stationary phases in reversedphase chromatography (RPC): (1) Phenyl-, (2) C4-, (3) C8-,(4) C18-, (5) cyano- and (6) diol-phases.

Page 71: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 55

electrons upon pyrolysis in the hydrogen–air flame,and thus enable an improved conduction of electric-ity. To a so-called collector located near the flame, apolarizing voltage is applied, which attracts gen-erated ions producing a current that is proportionalto the amount of sample being burned after elutionfrom the GC column. This particular current issensed by an electrometer, converted to a digital sig-nal, and plotted as peak in a chromatogram.

nitrogen phosphorus detector

The nitrogen phosphorus detector (NPD) is a vari-ant of the FID. The main difference is a glass or ceramic bead that is located right above the jet ofthe hydrogen–air plasma. A low hydrogen/air ratiocan not sustain a flame, resulting in decreasedhydrocarbon ionization. But the alkali ions on thebead surface facilitate the ionization of nitrogen- orphosphorus-containing compounds, thus favoringthe detection of those compounds while suppress-ing the abundance of other, mainly hydrocarbon-based substances.

ultraviolet absorbance detector

While FID and NPD can only be interfaced to GC,the UV absorbance detector has proven to be one of

the most popular analyzers for high performanceLC since the late 1960s. The principle is based onconventional spectrophotometry and the Beer–Lambert law:

Log I0/I = εcd

with I0 = intensity of the incident light, I = intensityof the transmitted light, ε = extinction coefficient, c = concentration of the absorbing analyte and d = path length of the cell. In practice, the eluent of a HPLC analysis is passed through a cell of fixedlength, a light source emitting in a distinct UV range(e.g. deuterium lamp, 190–600 nm) and, providinglight of defined intensity is focused on the cellthrough a monochromator, the intensity of thetransmitted light of selected wave length is detectedby means of photo diodes.

The detectors described so far enable the recogni-tion of compounds chromatographically separatedeither by GC or HPLC. The main drawback of theseanalyzers is the low specificity and correlated lackof detailed information on the analytes. As a con-sequence, instruments based on MS have becomethe first choice in doping control analysis beingemployed in combination with both GC and HPLC,complementary to the ‘traditional’ instrumentationutilizing FID, NPD and UV detectors. In order toidentify substances by MS, ionization is required,which can be accomplished by many different tech-niques in consideration of the respective chroma-tographic system. Nowadays, GC-MS frequentlyemploys techniques such as electron ionization andchemical ionization, while HPLC is mainly inter-faced to mass spectrometers by ESI, atmosphericpressure chemical ionization (APCI) or also atmo-spheric pressure photo ionization (APPI). In the following, we briefly describe the principles of elec-tron ionization and ESI as well as quadrupole andion trap analyzers, the most commonly used tools indoping control laboratories.

Ionization techniques

electron ionization

Electron ionization (EI) is widely used to generate positive ions of analytes separated by GC. Here,

Air

Vent

Collector

Hydrogen–air flame

GC column

H2

Fig. 5.6 Principal design of flame ionization detectors(FIDs).

Page 72: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

56 chapter 5

electrons emitted from a cathode, the so-calledfilament, are accelerated to an anode, and collide byorthogonal orientation within the ion source withcompounds eluting from the GC column as demon-strated in Fig. 5.7. The collision of electrons releasedwith commonly utilized 70 eV (potential betweenfilament and anode is set to 70 V) with analytes in anion source gives rise to a number of processes; forexample:

(1) AB + e– → AB+ + 2e–

(2) AB + e– → AB2+ + 3e–

(3) AB + e– → AB–

For positive ionization, the most important mechan-ism is (1) with the generation of a positively chargedmolecule (AB+) by liberation of an electron uponimpact of the accelerated electron released from thefilament. In addition, also two electrons can beremoved from the analyte AB giving rise to a doublycharged molecule AB2+ (2). Moreover, electrons canbe incorporated into the molecule AB resulting in anegatively charged analyte AB– (3). Besides theseexamples of effects of EI on compounds introducedinto an ion source by GC, the consequences of thisionization technique also have to be taken into con-sideration. As a major result of EI, highly energetic

radical cations are generated inclining to dissociateinto product ions, which are detected in the massselective analyzer giving rise to a characteristicmass spectrum of the respective substance. Theseproduct ions can be radicals as well as even-electronions, depending on rearrangement and eliminationprocesses.

electrospray ionization

The development of the comparably soft ioniza-tion technique utilizing electrospray was recentlyawarded with the Nobel Prize for chemistry in 2002.John Fenn was honored for fundamental researchregarding the ionization of macromolecules thatwas already introduced by Yamashita and Fenn in1984 (Yamashita & Fenn 1984b; Voyksner 1997).With this technique, liquids containing protonatedor deprotonated molecules are sprayed by means ofa capillary tip at high voltages (1 kV and higher),forming charged droplets that shrink by solventevaporation and repeated droplet disintegrationleading to very small and highly charged droplets.Finally, gas-phase ions are produced from thesevery small droplets as discussed in two differenttheories, namely charged residue model (Dole et al.1968) and ion evaporation (Iribarne & Thomson 1976).In Fig. 5.8, the main processes of ESI are presented.First, the penetration of imposed electric field intothe liquid of the capillary leads to an enrichment ofpositive charges at the surface of the liquid, causingdestabilization of the meniscus, formation of a coneand a so-called jet-emission of droplets bearing anexcess of positive ions. The charged droplets shrinkby solvent evaporation while the charge remainsconstant. Hence, an increase of the electrostaticrepulsion occurs, resulting in fission of the dropletsas they reach the Rayleigh stability limit. This phe-nomenon continues with ongoing evaporation ofsolvent until very small and highly charged dropletsare created. Finally, as proposed by Dole et al. (1968),only one ion remains in a singly charged droplet,and the evaporation of solvent gives rise to a gas-phase ion (charged residue model). Alternatively,the direct ion emission from droplets with a radiussmaller than 10 nm was postulated, also generatinggas-phase ions (ion evaporation) (Kebarle & Ho 1997).

Filament

Anode

e–

GC column

To massselectivedetector

Fig. 5.7 Schematic assembly of an ion source with electronionization.

Page 73: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 57

Mass analyzers

The ions generated from substances by various ion-ization techniques are analyzed by different massselective detectors such as quadrupole or ion trapinstruments. Both analyzers operate under highvacuum and can be interfaced to GC (and thus EI) aswell as LC (including ESI) by respective couplingsystems. As EI is performed under high vacuum, no special interface is necessary in order to control different pressure levels, but ESI operates at atmo-spheric pressure. Hence sophisticated vacuum barriers are present in modern LC-ESI-MS(/MS)systems maintaining the required reduced pressurein mass selective detectors.

quadrupole analyzers

A quadrupole mass spectrometer is composed byfour rods consisting of various materials; for exam-ple, fused silica coated with a gold layer. Opposingrods are connected while adjacent segments areelectrically isolated, as demonstrated in Fig. 5.9.Initially, ions that are generated in the ion source areaccelerated into the center of the quadrupole. Whileapplying alternating current (ac) to the segments,

positive or negative fields are established towardsthe centerline of the quadrupole and, thus, positiveions passing through the rods are pushed away withpositive and attracted with negative polarization.The extent of ion deflection is directly depending onthe applied voltage, its frequency (i.e. the durationof exposure to alternating fields) and the mass of ions. In addition, to one segment positive directcurrent (dc) is applied while the other segment isprovided negative dc. As a result, ions of a distinctmass to charge ratio can travel through the two-dimensional quadrupole field if the applied directcurrent and alternating current with a defined fre-quency are appropriate for stable oscillating moving.These parameters can be modified for an optimizedmass selection, enabling the isolation of a single ionor the recording of a full spectrum by scanning a dis-tinct range of mass/charge ratios over a given timeperiod (Budzikiewicz 1998; Lottspeich & Zorbas1998).

Ions isolated by quadrupole instruments can be subjected to further experiments as frequ-ently employed in triple stage quadrupole mass

Connected segment

Single rod

-U-Vcos2πft

U+Vcos2πft

Ions

Fig. 5.9 General assembly of a quadrupole mass selectiveanalyzer. Two opposing metal or gold-coated fused silicarods are electrically connected while adjacent rods areisolated. Ions are accelerated from the ion source into thecenter of the quadrupole analyzer and their trajectoriesare influenced by application of ac and dc voltage. Onlyions of defined mass/charge ratios pass through thequadrupole at distinct voltages, which enables massfiltering.

Capillary with solvents and analytes

Taylor-cone

Charged droplets

Orifice plate

High-voltage power supply

Fig. 5.8 Processes in positive electrospray ionization.Application of high voltage to a liquid-filled capillaryleads to enrichment of positive ions at the liquid surface,destabilization of the meniscus and generation of a conethat emits droplets containing an excess of positivecharges.

Page 74: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

58 chapter 5

spectrometers. Here, three quadrupoles are present,the first one of which is used to pick a single ionfrom the ion beam introduced into the mass spec-trometer. In the second quadrupole, the so-calledcollision cell, the isolated ion hits molecules of a collision gas (e.g. argon, nitrogen) and dissociateswith respect to the applied accelerating voltage andcollision gas pressure. The resulting product ionsare finally measured in a third quadrupole, either by full scan or selected ion monitoring, providinginformation on fragmentation processes as well asstructure of the analytes.

quadrupole ion trap analyzers

A frequently employed alternative to quadrupolemass spectrometers are (quadrupole) ion trap analyzers. Already in 1953, Paul and co-workersdescribed the applicability of a three-dimensionalquadrupole to ‘trap’ ions, and in the last decades,sophisticated ion trap mass spectrometers weredeveloped based on this invention (Louris et al.1987, 1989; Patterson et al. 2002). In Fig. 5.10, theprincipal composition of an ion trap instrument isshown, including two end-cap electrodes (one with

an ion inlet and one with an ion exit aperture) and aring electrode, in the center of which ions are storedby application of appropriate rf field establishedbetween the ring electrode and the two end-capelectrodes. Ions that are generated by an externalion source (e.g. EI or ESI) are transferred into the iontrap, which contains a gas (e.g. helium, argon) atapproximately 1 mtorr. A commonly employed gasis helium that damps the trajectories of ions to thecenter of the ion trap, and moreover it reduces thekinetic energy of ions through collisions favoringthe storage of the ions within the trap. The storage of ions of a broad range of mass/charge ratios is directly depending on the ac voltage applied tothe ring electrode. Hence, the consecutive ejection of ions enabling their mass selective detection isaccomplished by the so-called instability mode,which is based on the successive increase of the acvoltage applied to the ring electrode in combinationwith an ac voltage applied to the end-cap electrodescausing resonant motion. Ions of defined mass/charge ratios develop instable trajectories, are ejectedthrough the perforations of the end-cap electrode(ion exit) and detected with an electron multiplier.

In addition to an efficient scan operation mode,ion trap mass spectrometers offer possibilities ofMSn experiments. With the selective removal of ionsfrom the trap, storage of a precursor ion of interestand its resonant activation, collisionally activated dis-sociation (CAD) is obtained, giving rise to production spectra with a comparable amount of informa-tion as obtained with triple stage quadrupole MS/MS experiments. But an important advantage of theion trap technique is that trapping of ions allows theselection, storage and subsequent dissociation of a product ion generated by MS/MS experiments,namely MS3, providing information on the fragmen-tation pathway and composition of product ions,which is of great interest, in particular in ESI-MS.

Sample preparation and purificationstrategies

In doping controls, mainly urine specimens areobtained from athletes in order to be analyzedregarding prohibited compounds according to estab-lished anti-doping rules. In addition, also samples

End-cap electrode(ion entrance)

Ground

Dipolar rf

Aperture

End-cap electrode(ion exit)

Ringelectrode

Quadrupolar rf

Ions

He He

HeHe

Fig. 5.10 Principal design of an ion trap massspectrometer. The ring electrode is edged by two end-cap electrodes establishing an rf field upon voltageapplication that enables the storage of ions in the center of the ion trap.

Page 75: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 59

of whole blood, plasma/serum and hair are obtainedfor certain purposes of drug identification. In orderto accomplish utmost specificity and required sensit-ivity in analyses, sample pretreatment has become avery important part of doping controls, and variousstrategies are present to purify specimens from saltsand interfering compounds, and to isolate the targetanalyte(s) in particular.

Liquid–liquid extraction

One of the oldest and most frequently utilized sample preparation steps is liquid–liquid extraction(LLE). The very first publications on the identifica-tion of benzedrine (a racemic mixture of amphet-amine sulfate) and metabolites in human urineemployed LLE in order to determine excretion rates(Richter 1938). Here, urine samples were alkalizedby means of 2 N sodium hydroxide and extractedwith petroleum ether. In the following decades,numerous applications using LLE for the purifica-tion of biological fluids were presented, all of whichwere based on the same principle of adjusting pHand extraction of acidic, neutral or alkaline com-pounds (Keller & Ellenbogen 1952; Axelrod 1954;Beckett & Rowland 1965; Beckett & Wilkinson 1965;Kolb & Patt 1965; Cartoni & Cavalli 1968). Theknowledge about pI of analytes provides the optionof multiple extraction of specimens and/or re-extraction of compounds at various pH values intodifferent solvents. With the desire for more detailed,specific and sensitive analytical assays, LLE pro-cedures were improved and extended in order toremove coextracted interfering substances, andthese principles have been utilized for up-to-datescreening and confirmation methods in doping control analysis (Donike et al. 1970; Spyridaki et al.2001; Van Eenoo et al. 2001; Thevis et al. 2003b). For instance, stimulants such as amphetamines,ephedrines and related derivatives are commonlyextracted from urinary matrices into ethers underalkaline conditions, and the organic layer is con-centrated and subsequently analyzed by GC-MSand/or GC-NPD. Moreover, LLE is employed forthe analysis of corticosteroids (Fluri et al. 2001),diuretics, β-receptor blocking agents and anabolicsteroids under various conditions (i.e. salt addit-

ives, organic solvents and pH values) (Geyer et al.1997; Thevis et al. 2001; Deventer et al. 2002).

A very powerful tool turned out to be the so-called re-extraction, which was already employed in 1952 (Keller & Ellenbogen 1952) and applied todoping control analysis in order to enhance purityof samples to be analyzed. In particular, the extrac-tion of analytes under alkaline conditions into anorganic solvent (e.g. diethyl ether) and subsequentretransfer of components of interest from theorganic into an acidified aqueous layer (e.g. 0.06 N HCl) proved to be very useful for confirmationmeasurements, for instance for the β2-agonist clen-buterol, by highly efficient reduction of biologicalbackground (Sigmund et al. 1998).

Solid-phase extraction

For a series of compounds, an alternative approachto purify biological samples and concentrate ana-lytes is solid-phase extraction (SPE). Based on various adsorbing materials such as polystyrene,C18 or ion exchange phases, several screening andconfirmation procedures in doping control analysishave been established. In 1968, Bradlow demon-strated the possibility to extract steroid conjugateswith neutral resins (Bradlow 1968), and furtherimprovements in material and methods have pro-vided tools for the development of procedures fordoping control purposes, for example, for anabolicsteroids (Donike et al. 1984) and diuretics (Thieme et al. 2001). A major advantage of SPE is the feas-ibility of full automation of sample extraction, in-cluding conditioning of cartridges, sample loading,washing and subsequent elution. Moreover, derivat-ization of analytes can be accomplished during SPE(Lisi et al. 1991).

Immunoaffinity chromatography

A more recently introduced way of analyte isolationfrom biological matrices is immunoaffinity chro-matography (IAC). In general, mono- or polyclonalantibodies able to bind non-covalently to distinctparts of target molecules are linked to a carrier suchas spherical agarose particles. The resulting ‘gel’ isplaced in a column bearing a frit, urine or plasma is

Page 76: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

60 chapter 5

loaded onto the column and passed through the gel.The carrier-linked antibodies recognise target ana-lytes as they flow through the column and capturethese compounds by generating non-covalent com-plexes, while other, non-antigenic substances miss-ing the antibody–antigen reaction, are eluted. Undervarious conditions, the non-covalent complex canbe broken without harming the antibody or analyteof interest and the purified antigen is analyzed, forexample by GC-MS, basically without any inter-

fering signals. The principal production of an IACcolumn and its use is shown in Scheme 5.5. Com-monly employed carriers are commercially avail-able agarose-beads. The surface of these beads isactivated in order to chemically couple peptides or proteins, which is accomplished by various techniques. One of the most frequently employedmethods is cyanogen bromide (CNBr) activationthat was introduced in 1967 by Axen et al. (1967).Under alkaline conditions, CNBr introduces cyanate

Agarose

Cyclic imidocarbonateslightly reactive

Substitutedimidocarbonate

Isourea derivative

Non-covalently bondantigen (target compound)Target analyte

Mixture ofcompounds

Covalently bondantibodySample

IAC column

Gel

Frit

Agarose-bead

Elution

Target compound

Glass tube

Cyanate esterhighly reactive

Scheme 5.5 Activation of agarose and subsequent immobilization of antibodies. IAC, immunoaffinity chromatography.

Page 77: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 61

esters and imidocarbonates into the matrix ofagarose by reaction with its surface hydroxyl func-tions to which antibodies are chemically connectedas isourea or substituted imidocarbonate derivat-ives (Hermanson et al. 1992). The beads bearingcovalently fixed antibodies are placed in a column,which recognise and retain only specific analytes ordefined classes of compounds present in biologicalfluid. With appropriate eluents, for example mix-tures of organic solvents and water, the capturedanalytes are released from the antibodies, concent-rated and analyzed by conventional techniquessuch as MS (Schänzer et al. 1996; Machnik et al. 1999). The reduction of biological backgroundresults in improved signal/noise ratios, and thus inbetter detection limits of GC-MS and LC-MS/MSinstruments.

Derivatization techniques

GC-MS is one of the primary tools for screening andconfirmational analyzes in doping control laborator-ies. These systems provide specificity and sensitiv-ity for numerous compounds that are prohibitedaccording to the list of banned substances of IOCand WADA. But a principle requirement to identifydrugs by means of GC-MS is volatility. As manyanalytes are heavy volatile, for example β2-agonists,various diuretics and β-receptor blocking agents,they are derivatized to more volatile analogs bymeans of different reagents. Here, hydroxyl and primary or secondary amino functions are targetgroups because of their ability of hydrophilic inter-

actions causing decreased volatility. One of the firstmodifications of analytes was accomplished byacetylation utilizing acetic anhydride. The resultingmolecules have to be purified from remainders ofacetic acid and acetic anhydride, commonly achievedby LLE. Because of this laborious and time-consum-ing way of preparing compounds for GC-MS ana-lysis, several derivatization agents were developed,preferably including trimethylsilyl (TMS) residues.There are for instance hexamethyldisilazane (HMDS),trimethylsilyl-imidazole (TMSImi) and N-methyl-N-trimethylsilyltrifluoroacetamide (MSTFA) (Donike1969), the latter one of which is nowadays the mostfrequently employed derivatization reagent, in par-ticular in combination with ammonium iodide and ethanethiol, which gives rise to in situ gener-ated trimethyliodosilane (TMIS), a highly reactivetrimethylsilylation reagent (Donike 1973; Donike & Zimmermann 1980). With TMIS, hydroxyl andamino groups are modified and also keto functionsare converted to their enol-TMS ethers, as demon-strated in Scheme 5.6. Several other powerfulreagents have been used that introduce trifluo-roacetyl (TFA)- or heptafluorobutyl (HFB) -residuesinto molecules, for example N,N-bistrifluoroaceta-mide, N-methyl-N-bistrifluoroacetamide (MBTFA)(Donike & Derenbach 1976) or N-methyl-N-bishep-tafluoroburtyramide (MBHFB), respectively. Thetrifluoroacetylation results in more stable derivat-ives of amino functions than trimethylsilylation,hence selective modification of analytes with TMSand TFA groups has also become possible, enablingthe gas chromatographic separation of stereoisomers

Scheme 5.6 Derivatization of doping-relevant analytes by means of trimethyliodosilane (TMIS).

Page 78: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

62 chapter 5

of stimulants such as ephedrines (Opfermann &Schänzer 1996; Thevis et al. 2003b).

Qualitative analysis of doping-relevantcompounds

For most of the prohibited compounds in elitesports, qualitative evidence of their presence inurine samples of athletes is sufficient for a positivetest result. As many assays for screening and con-firmation purposes are based on chromatographyinterfaced to MS, guidelines for the identification of substances by means of GC-MS or LC-MS(/MS)systems have been established. The identification ofa compound is accomplished if the relative abund-ances of a specified amount of characteristic ions(depending on MS techniques) agrees, with respectto allowed variations, with those obtained by ana-lysis of the corresponding reference material. Inaddition, retention times of signals observed inurine samples of athletes and in appropriatelyfortified reference urine specimens are comparedand may shift only within a defined time frame.Hence, knowledge about chromatographic and, inparticular, mass spectrometric properties of respect-ive analytes is of utmost importance in order tocharacterize and identify compounds in a complexmatrix such as urine. Numerous investigationsregarding the mass spectrometric behavior of per-formance enhancing or masking agents and theirdetection for doping control purposes have beenperformed (Masse et al. 1989; de Boer et al. 1991;Donike et al. 1995; Ayotte et al. 1996; Shackleton et al.1997; Bowers 1998; Aguilera et al. 1999; Thevis et al.2000, 2001, 2002, 2003a; Ventura et al. 2000; Leinonenet al. 2002), and the principles of some procedureswill be described in the following.

Anabolic steroids

Considering statistical data regarding conducteddoping controls and classes of abused drugs,anabolic steroids are the most frequently detectedprohibited compounds. In 2001, for example, morethan 40% of the banned substances identified by 25 IOC-accredited laboratories were related toanabolic agents. One representative of this category

is methyltestosterone, a derivative of testosteroneobtained by introduction of an additional methylgroup at C-17. Most anabolic steroids are subjects of extensive metabolism, giving rise to a series ofmetabolites, for instance by reduction of keto func-tions, oxidation of hydroxyl groups, introduction of hydroxyl residues and oxidation/reduction of carbon-carbon bonds of the steroid nucleus(Schänzer 1996). Following this phase-I metabol-ism, conjugation to glucuronides and/or sulfates isobserved as common phase-II modification prior to renal excretion. In Scheme 5.7, the principalmetabolism of methyltestosterone is demonstrated,generating 17α-methyl-5α-androstan-3α,17β-diol(I), 17α-methyl-5β-androstan-3α,17β-diol (II) andcorresponding glucuronic acid conjugates.

Commonly employed strategies to identify meta-bolites of anabolic steroids are based on the enzymatic hydrolysis of phase-II metabolites to thecorresponding phase-I metabolites, purification, con-centration, derivatization and subsequent GC-MSanalysis. For most anabolic steroid metabolites, noendogenous production in humans is possible,except for nandrolone, which will be discussedlater. Hence, in case of qualitative determination ofthese compounds in urine samples of athleteselected for doping controls, a positive test result will be reported. In Fig. 5.11a, the EI-mass spectrumof 17α-methyl-5α-androstan-3α,17β-diol after bis-trimethylsilylation is presented, and Fig. 5.11bshows a typical GC-MS chromatogram with char-acteristic ion traces of the methyltestosterone metab-olites found in a urine sample.

designer steroids

The problem of so-called designer steroids triggeredan avalanche in the sport as well as the scientificworld in October 2003 (Knight 2003) when the doping control laboratory of the University ofCalifornia, Los Angeles (UCLA) identified a com-pound related to gestrinone, a drug administered incases of endometriosis. Its hydrogenation at theethinyl residue at carbon 17 results in a steroid hor-mone termed tetrahydrogestrinone (THG), which canbe considered as an analog to the highly efficientanabolic steroid trenbolone, but the physiological

Page 79: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 63

effects and side effects of THG have never been clinically investigated. The commonly employedstrategy of doping control laboratories analyzingpharmaceutically produced and clinically testedremedies is obviously not sufficient to cope with thewillingness of some athletes to cheat and to risktheir health in order to win by a short head againstcompetitors. Owing to the fact that many drugscreening procedures are based on the comparisonof reference compounds to urine samples by meansof mass spectrometric techniques such as selectedion monitoring (SIM) or multiple reaction monitor-ing (MRM), unknown derivatives of remedies, such

as THG, are invested with invisibility for these conventional methods. Hence, more flexible assayshave been established enabling the detection ofknown as well as unknown drugs by common struc-tures such as a principal steroid nucleus, which ispossible in particular by means of modern LC-MS/MS systems.

Endogenous steroids

While the administration of anabolic steroids causesthe presence of metabolites normally not occur-ring in human urine samples, doping by means of

450400350300250

m/z Time (min)

200150100

73143

TMSO

105213228255

270318 360 435

H

OTMS

CH3

50

(a) (b)

100

80

60

40

20

0

Rel

ativ

e ab

un

dan

ce (

%)

0 11.90

m/z 435

m/z 255

m/z 143

(I)

(II)

12.00 12.10 12.20 12.30 12.40 12.50 12.60 12.70

18 000 16 000 14 000 12 000 10 000

8000 6000 4000 2000

Ab

un

dan

ce

Scheme 5.7 Metabolism of methyltestosterone to 17α-methyl-5α-androstan-3α,17β-diol (I), 17α-methyl-5β-androstan-3α,17β-diol (II) and their glucuronides.

Fig. 5.11 (a) Electron ionization-mass spectrum of the bis-trimethylsilylated 17α-methyl-5α-androstan-3α,17β-diol (mol. wt. = 450); (b) gas chromatography-mass spectrometry (GC-MS) chromatogram of characteristic ion traces of bis-trimethylsilylated 17α-methyl-5α-androstan-3α,17β-diol (I) and 17α-methyl-5β-androstan-3α,17β-diol (II).

Page 80: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

64 chapter 5

testosterone is more difficult to uncover as it is alsoproduced endogenously. Here, different approacheshave been established to determine applications oftestosterone, and the two most frequently utilizedtools are the testosterone/epitestosterone (T/E)ratio and the so-called isotope ratio mass spectro-metry (IRMS) of carbons. The profile of endogenoussteroids varies under numerous influences (Geyer et al. 1996), but the ratio of T and E has proven to be a reliable parameter that indicates abuse of testo-sterone as the production of epitestosterone is inde-pendent from testosterone. A threshold ratio of six is utilized that does not immediately entail a positive test result but triggers follow-up studies totest for an abnormal but naturally elevated testo-sterone concentration.

With the availability of IRMS instruments, num-erous investigations have been published demon-strating the possibility to distinguish betweenendogenously generated and chemically synthes-ized testosterone by MS. Naturally produced tes-tosterone comprises different 13C/12C ratios thanchemically synthesized analogs that are used formedical supplementation. By means of GC, com-bustion of eluting analytes and mass spectrometricmeasurement of resulting carbon dioxide providesinformation about the origin of testosterone by different contents of 13C and 12C (Horning et al. 1998;Aguilera et al. 1999).

Diuretics and ββ2-agonists

Two representatives of classes of compounds fre-quently analyzed by means of LC-MS(/MS) arediuretics and β2-agonists. In particular the categoryof diuretics demonstrates the chemical heterogene-ity of drugs administered for comparable or ident-ical purposes. Here, primarily negative ionization is performed (Thieme et al. 2001; Thevis et al. 2002,2003a) owing to the acidity of drugs belonging to the group of diuretics, but few compounds such astriamterene require positive ionization. For β2-agonists, protonation of analytes and detection of positively charged molecules is the method ofchoice. As described in Analytical techniques above,ion sources interfacing LC to MS mainly generateprotonated or deprotonated molecules without anyconsiderable fragmentation. Thus, structure infor-

mation about analytes, specificity and selectivity of mass analyzers are provided by CAD of ionizeddrugs and analysis of derived fragments. Thisrequires the knowledge of proton affinity and dis-sociation behavior after efficient activation of targetmolecules by CAD, which differs significantly fromfragmentation routes observed with EI. For diuret-ics as well as for the majority of β2-agonists (exceptfor salbutamol, see Quantitative analysis of pro-hibited drugs below), qualitative analysis of thesedrugs is sufficient, and typical ESI product ion spectra of epithiazide and fenoterol are presented inFig. 5.12. Commonly, extracted ion chromatogramsenable the sensitive detection of these compoundsin biological matrix, and confirmation of their pres-ence is obtained by comparison of relative abund-ances of product ions. In 2001, approximately 17.5%of the banned substances detected in 25 IOC-accred-ited laboratories in doping control samples weredrugs related to β2-agonists, and 5% were classifiedas diuretics.

Quantitative analysis of prohibited drugs

For several compounds, including stimulants suchas ephedrines, metabolites of anabolic steroids suchas nandrolone, and β2-agonists such as salbutamol,threshold levels have been established, which arethe basis of the decision as to whether a sample isreported positive or negative. Various reasons aregiven for this regulation. Ephedrines are ingredientsof many remedies frequently used in cold therapy,hence their use is legal in the sense of anti-dopingrules as long as their urinary concentration does notexceed 5, 10, or 25 µg·mL–1 for the derivatives nore-phedrine, ephedrine and pseudoephedrine, respect-ively. Salbutamol, a sympathomimetic agent, is one of four permitted β2-agonists (in addition to salmeterol, terbutaline and formoterol), as long asthe application is conducted via inhalation. As thedifferentiation between orally administered pillsand pressurized aerosol dosage is very complicated,the presence of salbutamol in competition samplesis reported to the corresponding federation if theurinary level exceeds 100 ng·mL–1, and in out-of-competition testing a threshold of 1 µg·mL–1 hasbeen established owing to anabolic effects observed

Page 81: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 65

with some sympathomimetics if administered indoses much higher than therapeutically used. Thepresence of the nandrolone metabolite 5α-estran-3α-ol-17-one (norandrosterone) in urine samples ofelite athletes is observed to certain extents based onendogenous production. Thus, also here thresholdvalues have been introduced with 2 ng·mL–1 formale and 5 ng·mL–1 for female athletes. Numerousstudies were performed in order to substantiatethese levels, and various ‘parameters’ influencingthe endogenous generation of this metabolite weretaken into consideration, for example, high physio-logical stress or pregnancy, which cause signific-antly elevated concentrations in urine specimens.Quantitation of these substances is accomplished bycalibration curves utilizing regular sample prepara-tion procedures and appropriate internal standardswith comparable or identical physicochemical prop-erties (Schänzer & Donike 1995).

Summary

Doping control analysis of low molecular weightdrugs is generally based on chromatographic andmass spectrometric techniques that enable thedetection and identification of remedies and theirmetabolites in body fluids such as urine and blood.While early procedures employed mainly GC, vari-ous analyzers such as FIDs and NPDs, as well as MS, recently published assays utilize primarily LCinterfaced to MS by means of atmospheric pressure

ionization techniques due to a faster sample prepara-tion as no derivatization of analytes is required. Inaddition, more flexible mass spectrometric experi-ments to determine and characterize known thera-peutics and also unknown designer drugs havebecome possible by frequently employed triplequadrupole or ion trap analyzers supporting thefight against doping and the illegal use of drugs.The range of substances important for doping con-trol analysis has changed ever since lists of pro-hibited compounds and methods of doping haveexisted, and laboratories permanently expand ormodify analytical procedures within this dynamicprocess in order to improve performance regardingsensitivity, specificity and flexibility to limit the mis-use of drugs in sport as well as to protect athletesfrom false suspicion. Here, new developments con-cerning high speed and high resolution chromato-graphy, high resolution and high sensitivity MS aswell as modern ionization techniques provide valu-able tools for analytical laboratories to obtain evenmore detailed information on the administeredtherapeutics, for example their chemical structuresand metabolism, and enable extended time framesfor the determination of drug abuse. As many therapeutics, such as anabolic steroids, are applied during out-of-competition periods but preserveperformance-enhancing effects for several weeks,doping control analysis requires in- as well as out-of-competition tests and the utmost specificity andsensitivity of analytical procedures.

300280260240220200180160140

152

HO

OHOH

N H

N H

HO

135

107

164 170

286 304 62 95

126

150169

228260 279

341

310

300

269

205

78Cl

S

S CF3

NH

OO

364 384

404

424

120100

H3CH2NO2S

806040

(a) (b)m/z m/z

100 90 80 70 60 50 40 30 20 10 0

Rel

ativ

e ab

un

dan

ce (

%)

400350200 250 30015010050

100 90 80 70 60 50 40 30 20 10 0

Rel

ativ

e ab

un

dan

ce (

%)

m/z 107m/z 135

Fig. 5.12 (a) Electrospray ionization (ESI) product ion spectrum of the β2-agonist fenoterol (mol. wt. = 303); (b) ESI production spectrum of the diuretic agent epithiazide (mol. wt. = 425).

Page 82: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

66 chapter 5

References

Aguilera, R., Catlin, D.H., Becchi, M. et al.(1999) Screening urine for exogenoustestosterone by isotope ratio massspectrometric analysis of onepregnanediol and two androstanediols.Journal of Chromatography B 727, 95–105.

Ariëns, E.J. (1965) General andpharmacological aspects of doping. In: Doping (de Schaepdryver, A. &Hebbelink, M., eds.). Pergamon Press,Oxford: 27–50.

Axelrod, J. (1954) Studies onsympathomimetic amines. II. Thebiotransformation and physiologicaldisposition of d-amphetamine, d-p-hydroxyamphetamine and d-methamphetamine. Journal ofPharmacology and ExperimentalTherapeutics 110, 315–326.

Axen, R., Porath, J. & Ernback, S. (1967)Chemical coupling of peptides andproteins to polysaccharides by means ofcyanogen halides. Nature 24, 1302–1304.

Ayotte, C., Goudreault, D. & Charlebois,A. (1996) Testing for natural andsynthetic anabolic agents in humanurine. Journal of Chromatography B 687,3–25.

Beckett, A.H. & Rowland, M. (1965)Determination and identification ofamphetamine in urine. Journal ofPharmacy and Pharmacology 17, 59–60

Beckett, A.H. & Wilkinson, G.R. (1965)Identification and determination ofephedrine and its congeners in urine bygas chromatography. Journal of Pharmacyand Pharmacology 17 (Suppl.), S104–S106.

Beckett, A.H., Tucker, G.T. & Moffat, A.C.(1967) Routine detection andidentification in urine of stimulants andother drugs, some of which may be usedto modify performance in sport. Journalof Pharmacy and Pharmacology 19,273–294.

de Boer, D., Gainza Bernal, M.E., vanOoyen, R.D. & Maes, R.A. (1991) Theanalysis of trenbolone and the humanurinary metabolites of trenboloneacetate by gas chromatography/massspectrometry and gas chromatography/tandem mass spectrometry. BiologicalMass Spectrometry 20, 459–466.

Bowers, L.D. (1998) Athletic drug testing.Clinics in Sports Medicine 17, 299–318.

Bradlow, H.L. (1968) Extraction of steroidconjugates with a neutral resin. Steroids11, 265–272.

Budzikiewicz, H. (1998)Massenspektrometrie. Wiley–VCH, New York.

Burstin, S. (1963) La Lucha contra elDopage. Revista de Derecho Sportiva 3,583–636.

Cartoni, G.P. & Cavalli, A. (1968) Detectionof doping by thin-layer and gaschromatography. Journal ofChromatography 37, 158–161.

Deventer, K., Delbeke, F.T., Roels, K. &Van Eenoo, P. (2002) Screening for 18diuretics and probenecid in dopinganalysis by liquid chromatography-tandem mass spectrometry. BiomedicalChromatography 16, 529–535.

Dole, M., Mack, L.L., Hines, R.L. et al.(1968) Molecular beams of macroions.Journal of Chemical Physics 49, 2240–2249.

Donike, M. (1969) N-Methyl-N-trimethylsilyl-trifluoracetamid, einneues Silylierungsmittel aus der Reiheder silylierten Amide. Journal ofChromatography 42, 103–104.

Donike, M. (1973) Acylierung mitBis(Acylamiden): N-Methyl-bis(trifluoracetamid) undBis(trifluoracetamid), zwei neueReagenzien zur Trifluoracetylierung.Journal of Chromatography 78, 273–279.

Donike, M. & Derenbach, J. (1976) Dieselektive Derivatisierung unterkontrollierten Bedingungen: Ein Wegzum Spuren-Nachweis von Aminen.Zeitschrift Analytische Chemie 279,128–129.

Donike, M. & Zimmermann, J. (1980) Zur Darstellung von Trimethylsilyl-,Triethylsilyl- und tert.-Butyldimethylsilyl-enoläthern von Ketosteroiden für gas-chromatographische undmassenspektrometrischeUntersuchungen. Journal ofChromatography 202, 483–486.

Donike, M., Jaenicke L., Stratmann, D. & Hollmann, W. (1970) Gaschromatographic detection of nitrogen-containing drugs in aqueous solutionsby means of the nitrogen detector.Journal of Chromatography 52, 237–250.

Donike, M., Zimmermann, J., Bärwald,K.R. et al. (1984) Routinebestimmungvon Anabolika im Harn. DeutscheZeitschrift für Sportmedizin 35, 14–24.

Donike, M., Ueki, M., Kuroda, Y. et al.(1995) Detection of dihydrotestosterone(dht) doping: alterations in the steroidprofile and reference ranges for dht andits 5 α-metabolites. Journal of SportsMedicine and Physical Fitness 35, 235–250.

Engelhardt, H., Müller, H. & Schön, U.(1985) The column. In: High Performance

Liquid Chromatography in Biochemistry(Henschen, A., Hupe, K.P., Lottspeich, F.& Voelter, W., eds.). VCHVerlagsgesellschaft, Weinheim: 17–78.

Fluri, K., Rivier, L., Dienes-Nagy, A. et al.(2001) Method for confirmation ofsynthetic corticosteroids in doping urinesamples by liquid chromatography-electrospray ionization massspectrometry. Journal of ChromatographyA 926, 87–95.

Geyer, H., Schänzer, W., Mareck-Engelke,U. & Donike, M. (1996) Factorsinfluencing the steroid profile. In:Proceedings of the 14th Cologne Workshopon Doping Analysis (Donike, M. Geyer,H., Gotzmann A. & Mareck-Engelke, U.,eds.). Sport & Buch Strauß, Cologne:95–114.

Geyer, H., Schänzer, W., Mareck-Engelke,U., Nolteernsting, E. & Opfermann, G.(1997) Screening procedure for anabolicsteroidsathe control of the hydrolysiswith deuterated androsteroneglucuronide and studies with directhydrolysis. In: Proceedings of the 15thCologne Workshop on Doping Analysis(Schänzer, W., Geyer, H., Gotzmann, A.& Mareck-Engelke, U., eds.). Sport &Buch Strauß, Cologne: 99–101.

Hermanson, G.T., Mallia, A.K. & Smith,P.K. (1992) Immobilized Affinity LigandTechniques. Academic Press, San Diego,CA.

Höher, J. & Troidl, H. (1995) Doping im Sport. Deutsche Zeitschrift fürSportmedizin 46, 270–282.

Horning, S., Geyer, H., Gotzmann, A.,Flenker, U. & Schänzer, W. (1998)Detection of exogenous steroids by13C/12C analysis. In: Proceedings of the16th Cologne Workshop on Doping Analysis (Schänzer, W., Geyer, H.,Gotzmann, A. & Mareck-Engelke, U.,eds.). Sport & Buch Strauß, Cologne:135–148.

International Olympic Committee (IOC)(2003) List of Prohibited Substances andMethods of Doping. IOC, Lausanne.

Iribarne, J.V. & Thomson, B.A. (1976) Onthe evaporation of small ions fromcharged droplets. Journal of ChemicalPhysics 64, 2287–2294.

Kebarle, P. & Ho, Y. (1997) On themechanism of electrospray massspectrometry. In: Electrospray IonizationMass Spectrometry—Fundamentals,Instrumentation and Applications (Cole,R.B., ed.). John Wiley & Sons, New York:3–63.

Page 83: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

doping analysis with gc-ms and lc-ms/ms 67

Keller, R.E. & Ellenbogen, W.C. (1952) The determination of d-amphetamine in body fluids. Journal of Pharmacologyand Experimental Therapeutics 106, 77–82.

Knight, J. (2003) Drugs bust revealsathletes’ secret steroid. Nature 425, 752(News).

Kolb, H. & Patt, P.W. (1965) Beitrag zum Arzneimittelnachweis inKörperflüssigkeiten durchGaschromatographie. Arzneimittel-Forschung 15, 924–927.

Lang, H.J. & Hropot, M. (1995) Discoveryand development of diuretic agents. In:Handbook of Experimental Pharmacology,vol. 117 (Greger, R.F., Knauf, H. &Mutschler, E., eds.). Springer-Verlag,Berlin: 141–172.

Leinonen, A., Kuuranne, T. & Kostiainen,R. (2002) Liquid chromatography/mass spectrometry in anabolic steroidanalysisaoptimization and comparisonof three ionization techniques:electrospray ionization, atmosphericpressure chemical ionization andatmospheric pressure photoionization.Journal of Mass Spectrometry 37, 693–698.

Lisi, A.M., Trout, G.J. & Kaslauskas, R.(1991) Screening for diuretics in humanurine by gas chromatography-massspectrometry with derivatisation bydirect extractive alkylation. Journal ofChromatography 563, 257–270.

Lottspeich, F. & Zorbas, H. (1998)Bioanalytik. Spektrum AkademischerVerlag, Heidelberg.

Louris, J.N., Cooks, R.G., Syka, J.E.P. et al.(1987) Instrumentation, applications,and energy disposition in quadrupoleion-trap tandem mass spectrometry.Analytical Chemistry 59, 1677–1685.

Louris, J.N., Amy, J.W., Ridley, T.Y. &Cooks, R.G. (1989) Injection of ions into a quadrupole ion trap massspectrometer. International Journal ofMass Spectrometry and Ion Processes 88,97–111.

Machnik, M., Geyer, H., Horning, S. et al. (1999) Long-term detection ofclenbuterol in human scalp hair by gaschromatography-high-resolution massspectrometry. Journal of ChromatographyB 723, 147–155.

Masse, R., Ayotte, C., Bi, H.G. & Dugal, R.(1989) Studies on anabolic steroids. III.Detection and characterization ofstanozolol urinary metabolites inhumans by gas chromatography-massspectrometry. Journal of Chromatography497, 17–37.

Möhrke, W. & Ullrich, F. (1995)Metabolism of diuretics. In: Handbook of

Sigmund, G., Horning, S., Seinsch, I. &Schänzer, W. (1998) Confirmation ofclenbuterol. Proceedings of the 16thCologne Workshop on Doping Analysis(Schänzer, W., Geyer, H., Gotzmann, A.& Mareck-Engelke, U., eds.). Sport &Buch Strauß, Cologne: 129–133.

Spyridaki, M.H., Tsitsimpikou, C.J., Siskos,P.A. & Georgakopoulos, C.G. (2001)Determination of ephedrines in urine by gas chromatography-massspectrometry. Journal of ChromatographyB 758, 311–314.

Stallion, A., Zhang, F.S., Chance, W.T.,Foley-Nelson, T. & Fischer, J.E. (1991)Reversal of cancer cachexia in rats bycimaterol and supplemental nutrition.Surgery 110, 678–684.

Thevis, M., Opfermann, G. & Schänzer, W.(2000) Detection of the plasma volumeexpander hydroxyethyl starch in humanurine. Journal of Chromatography B 744,345–350.

Thevis, M., Opfermann, G. & Schänzer, W.(2001) High speed determination of β-receptor blocking agents in human urineby liquid chromatography/tandemmass spectrometry. BiomedicalChromatography 15, 393–402.

Thevis, M., Schmickler, H. & Schänzer, W.(2002). Mass spectrometric behavior of thiazide-based diuretics afterelectrospray ionization and collision-induced dissociation. AnalyticalChemistry 74, 3802–3808.

Thevis, M., Schmickler, H. & Schänzer, W.(2003a). Effect of the location ofhydrogen abstraction on thefragmentation of diuretics in negativeelectrospray ionization massspectrometry. Journal of the AmericanSociety for Mass Spectrometry 14, 658–670.

Thevis, M., Sigmund, G. & Schänzer, W.(2003b) Confirmation ofephedrinesacomparison between GC-MS and LC-MS/MS. In: Proceedings of the 21st Cologne Workshop on DopingAnalysis (Schänzer, W., Geyer, H.,Gotzmann, A. & Mareck-Engelke, U.,eds.). Sport & Buch Strauß, Cologne: 303–307.

Thieme, D., Grosse, J., Lang, R., Mueller,R.K. & Wahl, A. (2001) Screening,confirmation and quantification ofdiuretics in urine for doping controlanalysis by high-performance liquidchromatography-atmospheric pressureionization tandem mass spectrometry.Journal of Chromatography A 757, 49–57.

Van Eenoo, P., Delbeke, F.T., Roels, K. &De Backer, P. (2001) Simultaneous

Experimental Pharmacology , vol. 117(Greger, R.F., Knauf, H. & Mutschler, E.,eds.). Springer-Verlag, Berlin: 173–200.

Morgan, C.E. (1957) Drug administrationto racing animals. Journal of the AmericanVeterinary Medical Association 130,240–243.

Opfermann, G. & Schänzer, W. (1996)Trimethylsilylationaaspects forderivatisation. In: Proceedings of the 14th Cologne Workshop on Doping Analysis (Schänzer, W., Geyer, H.,Gotzmann, A. & Mareck-Engelke, U.,eds.). Sport & Buch Strauß, Cologne:247–252.

Patterson, G.E., Guymon, A.J., Riter, L.S. et al. (2002) Miniature cylindrical iontrap mass spectrometer. AnalyticalChemistry 74, 6145–6153.

Prokop, L. (1970) The struggle againstdoping and its history. Journal of Sports Medicine and Physical Fitness 10,45–48.

Prokop, L. (2002) Zur Geschichte desDopings. Sportärztewoche, December 1–7,Kaprun.

Reeds, P.J., Hay, S.M., Dorward, P.M. &Palmer, R.M. (1988) The effect of β-agonists and antagonists on musclegrowth and body composition of youngrats (Rattus sp.). Comparative Biochemistryand Physiology C 89, 337–341.

Richter, D. (1938) A colour reaction forbenzedrine. Lancet 238, 1275.

Schänzer, W. (1996) Metabolism ofanabolic androgenic steroids. ClinicalChemistry 42, 1001–1020.

Schänzer, W. & Donike, M. (1995)Synthesis of deuterated steroids forGC/MS quantification of endogenoussteroids. In: Proceedings of the 12thCologne Workshop on Dope Analysis(Donike, M., ed.). Sport & Buch Strauß,Cologne: 93–112.

Schänzer, W., Delahaut, P., Geyer, H.,Machnik, M. & Horning, S. (1996) Long-term detection and identification ofmetandienone and stanozolol abuse inathletes by gas chromatography-high-resolution mass spectrometry. Journal ofChromatography B 687, 93–108.

Scott, R.P.W. (1982) Microbore columns inliquid chromatography. In: Techniques inLiquid Chromatography (Simpson, C.F.,ed.). John Wiley & Sons, Chichester:79–96.

Shackleton, C.H., Phillips, A., Chang, T. & Li, Y. (1997) Confirming testosteroneadministration by isotope ratio massspectrometric analysis of urinaryandrostanediols. Steroids 62,379–387.

Page 84: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

68 chapter 5

quantitation of ephedrines in urine by gas chromatography-nitrogen-phosphorus detection for doping controlpurposes. Journal of Chromatography B760, 255–261.

Venerando, A. (1963) Pathologia deldoping e mezzi di controllo. Medicinadello Sport 3, 945–959.

Ventura, R., Segura, J., Berges, R. et al.(2000) Distinction of inhaled and oralsalbutamol by urine analysis usingconventional screening procedures fordoping control. Therapeutic DrugMonitoring 22, 277–282.

Vidic, E. (1956) Eine Methode zur Identifizierung

vol. 2 (Emmet, P.H., ed.). Reinhold, New York: 105–165.

Wilhelm, M. & deStevens, G. (1976)Antihypertensive agents. Progress inDrug Research. 20, 197–259.

www.g-o.de/kap4/40gboo91.thm (06-2003)

Yamashita, M. & Fenn, J.B. (1984a)Electrospray ion source. Anothervariation on the free-jet theme. Journal of Chemical Physics 88, 4451–4459.

Yamashita, M. & Fenn, J.B. (1984b)Negative ion production with theelectrospray ion source. Journal ofChemical Physics 88, 4671–4675.

papierchromatographisch isolierterArzneistoffe. Archives of Toxicology 16,63–73.

Voyksner, R.D. (1997) Combining liquidchromatography with electrospray massspectrometry. In: Electrospray IonizationMass Spectrometry—Fundamentals,Instrumentation and Applications (Cole,R.B., ed.). John Wiley & Sons, New York:323–341.

Wagner, J.C. (1989) Abuse of drugs used toenhance athletic performance. AmericanJournal of Hospital Pharmacy 46,2059–2067.

Wheeler, A. (1955) Reaction rates andselectivity in catalyst pores. In: Catalysis,

Page 85: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

69

Abstract

The reproductive axis is largely stable in the face of intensive physical training in healthy men andwomen. However, altered reproductive-hormoneoutflow is associated with any of numerous dopingprotocols, exhaustive or non-escalating trainingschedules, concomitant weight loss, psychosocialstress, inadequate caloric intake for workload andsustained strenuous exertion in adolescence. Expo-sure to anabolic steroids induces loss of menstrualcyclicity in women, a decrease in spermatogenesisin men, and a significant reduction in protective(high-density lipoprotein) cholesterol concentra-tions in both genders. More subtle endocrine andmetabolic adaptations are detectable in the absenceof the foregoing risk factors, but their medicalsignificance is not established. For example, one longitudinal investigation showed that the onlydetectable effect of supervised long-distance run-ning training to complete a marathon in healthyyoung women is slight abbreviation of the post-ovulatory (luteal) phase of the menstrual cyclewithin the normal range. Cross-sectionally basedreports describe an array of reproductive abnorm-alities in athletes, but such data are confounded by one or more known comorbid factors. Accord-ingly, supervised, graded, non-exhaustive, voluntarystrenuous exercise in healthy adults maintaining anadequate caloric intake has minimal potential foradverse reproductive consequences.

Introduction

Critical assessment of the impact of vigorous exer-cise training and strenuous sports engagements on the endocrine system is a recent medical accom-plishment. Indeed, the majority of earlier clinicalreports cited in textbooks or forecast in contempor-ary advertisements of health-related supplementsare flawed by significantly confounding issues,which render definitive interpretation impossible.Scientific requirements for valid inference currentlyinclude (non-exclusively): (i) prospective evaluationof demonstrably normal healthy individuals; (ii)supervised, graded, escalating training schedules ofquantifiable intensity and duration; (iii) concurrentlongitudinally monitored gender and age-matchedcontrol cohorts without exercise intervention; (iv)verifiable adequacy of concomitant nutritional sup-port to match individual energy demand; (v) docu-mented absence of covert drug or hormone use(doping); (vi) baseline and anterograde assessmentof perceived psychosocial stress; and (vii) compre-hensive family history to identify genetic factorsknown to predict reproductive disorders in the gen-eral population. At present, no single study fulfillsthe foregoing ensemble expectations. More subtleunderstanding of metabolic and endocrine implica-tions of vigorous and competitive sports will thusrequire further clinical and physiological studies.

Principles of reproductive physiology

Contemporary knowledge of human reproduct-ive physiology embraces an integrative view of

Chapter 6

The Reproductive Axis

JOHANNES D. VELDHUIS AND ARTHUR L. WELTMAN

Page 86: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

70 chapter 6

hormonal signal exchange among the central nerv-ous system (hypothalamus), anterior pituitary gland(gonadotrope cells) and gonad (testis and ovary)(Fig. 6.1). The foregoing set of neuroendocrineglands (the reproductive axis) is linked to signalsassociated with pubertal development, somaticgrowth, body composition, stress responses, appetite,energy expenditure and insulin action (Urban et al.1988; Evans et al. 1992; Giustina & Veldhuis 1998).An integrative perspective of the reproductive axisis necessary to reconstruct the pathogenesis of endo-crine disturbances and discern the mechanisms of normal adaptations to exercise. For example,

adequate caloric repletion of expended energy is aminimal (necessary, but not sufficient) prerequisitefor successful reproductive function during strenu-ous training (Loucks 2003).

Endocrine adaptations to physical exertion aretransduced via multiple levels of non-exclusive control: (a) central-neural and blood-borne inputs to the hypothalamus; (b) hypothalamic integrationto yield convergent signal outflow to the anteriorpituitary gland; (c) feedback to the hypothalamusvia core body temperature, inflammatory mediators,tissue-derived metabolites (e.g. lactate, free fattyacids) and secreted products of target cells (insulin-

Neuroanatomic locus Primary messenger

Sex steroidsCNS

LH

LH

Time

HypothalamusPortalmicrovasculature

Pituitarygland

Gonad

Sex steroid-responsive tissues

(+) GnRH

(–)

(–)

(–)

(+)

LH, FSH, prolactin,sex steroids

Ovary: estrogen, progesteroneTestis: testosteroneGametogenesis

Circulation

Fig. 6.1 Schematized illustration of basic elements maintaining homeostasis of the male and female reproductive axes.Signals arising in the central nervous system and peripheral tissues converge on an ensemble of about 1200 hypothalamicgonadotropin-releasing hormone (GnRH) -secreting neurons. Synchrony of neuronal excitation releases pulses of GnRHinto a microvascular portal-venous system (solid arrows [+]), which connects the mediobasal hypothalamus to thepituitary gland. GnRH drives transcription, translation, processing and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) by gonadotrope cells. The general circulation delivers LH to gonadal (ovarian or testicular)compartments. LH stimulates the synthesis and secretion of testosterone (and thereby estradiol) in men and women, andprogesterone in women after ovulation. In women, FSH promotes Graafian follicle development, oocyte fertilizability andovarian responsiveness to LH. In men, FSH synergizes with LH-stimulated intratesticular testosterone to promote earlystages of spermatogenesis in seminiferous tubules. Sex steroids act on peripheral target tissues, such as muscle, fat, breast,bone, skin, sexual organs, the hypothalamus and pituitary gland (open arrow [–]). In women, estrogen and progesteroneexert sequential feedback (inhibition) and feedforward (enhancement), which cycle mediates the preovulatory LH surge.The same sex hormones stimulate uterine endometrial growth and maturation, which are necessary for implantation of a(fertilized) blastocyst in pregnancy.

Page 87: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the reproductive axis 71

like growth factor stimulated in the liver by growthhormone); (d) exogenous substrates (glucose, fatand amino acids); and (e) stress-adaptive thyroidaland adrenal hormones (e.g. thyroxine, cortisol andepinephrine). This minimal network of interactionsgoverns hypothalamic stimulation of gonadotropecells by intermittent (burst-like) release of the deca-peptide, gonadotropin-releasing hormone (GnRH).GnRH induces (feeds forward on) the pituitary syn-thesis and secretion of luteinizing hormone (LH)and follicle-stimulating hormone (FSH). These twoproteins are key effectors of the ovary and testis. Thesize and time-pattern of GnRH pulses jointly deter-mine the amounts of LH and FSH secreted (Urban et al. 1988; Evans et al. 1992; Veldhuis 1999). LHdrives sex-steroid production (below). FSH controlssperm and egg development and the production ofa feedback protein, inhibin. No clinically significantalterations in FSH or inhibin secretion have beenattributable to physical training (Veldhuis et al.1998). This statement is also true of prolactin, a lacta-tional hormone that increases transiently with about of exercise and in response to diverse psycho-logical and physical stressors.

In men, LH acts on testicular Leydig cells to pro-mote the synthesis and secretion of 4–6 mg oftestosterone each day (Urban et al. 1988). In women,LH stimulates ovarian theca and postovulatoryluteal cells to secrete about 0.15 mg of testosterone (a precursor of estrogens) and 10–20 mg of proges-terone per day (Evans et al. 1992). Testosterone, estro-gen and progesterone reversibly suppress (feedback on) the hypothalamo–pituitary unit, therebysupervising intermittent secretion of GnRH, LH andFSH (Veldhuis 1999). Time-delimited inhibition bysex-steroid hormones is a fundamental regulatorymechanism that preserves reproductive homeostasis.In general, homeostasis is the aggregate outcome of reciprocal feedforward (stimulatory) and feed-back (repressive) interactions that enforce repeated,reversible incremental adjustments to maintain hormone concentrations within the physiologicalrange for age, gender and species. Autoregulation isanalogous to a cruise-control device, which holds a vehicle’s speed within the desired preset range.Cascades of signal exchange in healthy indivi-duals thereby ensure life-supporting concentrations

of substrates (e.g. glucose), metabolites (e.g. lac-tate) and hormones (sex steroids, cortisol, insulin,growth hormone, etc). Homeostasis requires de-terministic physiological mechanisms, low back-ground randomness, a normal genetic endowment,adequate psychosocial support and successfuladaptation to internal and external stress (Veldhuis1996).

Hypothalamo–pituitary–gonadalhomeostasis in strenuous physicaltraining

Longitudinal clinical studies

Few clinical investigations have entailed prospect-ive randomization of a demonstrably healthy cohortof volunteers into strata of unequal training intens-ity, duration and/or type. One longitudinal studyassigned young menstruating women randomly toeither low (sublactate threshold) or high (supralac-tate threshold) intensity physical training (Rogol etal. 1992). The program comprised supervised long-distance running for 18 months with the individualgoal of completing a marathon. Reproductive hor-mones were measured at baseline and after 1 year of gradually escalating training volume at the pre-assigned exercise intensity. LH, FSH, prolactin and estradiol did not change detectably in the low-intensity exercise cohort or in a concurrent longit-udinal control group (recreationally active women).Women who trained consistently above the indivi-dually determined lactate threshold for an identicaltotal running distance of > 500 miles (> 800 km)manifested a small (1.8-day) abbreviation of theluteal phase of the menstrual cycle with no evidentdifference in mean progesterone concentrations(Rogol et al. 1992). Shortening of luteal-phase lengthdid not exceed normal month-to-month differencesrecognized in healthy premenopausal women.

The importance of adequate nutritional intake to match total caloric expenditure is illustrated in arecent prospective randomized intervention. In this2-month-long study, women undertaking weightreduction during training were more likely todevelop disturbances in ovulatory or luteal-phasetiming than individuals training identically with

Page 88: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

72 chapter 6

FastingBaseline

160012008004000160012008004000160012008004000

8

6

4

2

0

160012008004000

16

12

8

4

0160012008004000

16

12

8

4

0160012008004000

16

12

8

4

0

160012008004000

20

16

12

8

4

016001200800

Time (min)

4000

20

16

12

8

4

0160012008004000

20

16

12

8

4

0

GnRH + Fasting8

6

4

2

0

8

6

4

2

0

Seru

m L

H c

on

cen

trat

ion

(IU

·L–1

)

Fig. 6.2 Short-term fasting suppresses, and gonadotropin-releasing hormone (GnRH) infusion restores, pulsatileluteinizing hormone (LH) release in healthy young men. Profiles are shown in three individuals (top, middle, bottom). LH concentrations were sampled every 10 min for 24 h. The triptych illustrates the fed session (left); day 3 of fasting withsaline injections (middle); and day 3 of fasting accompanied by intravenous injection of synthetic GnRH (100 ng·kg–1

every 90 min) (right). Repeated GnRH pulses restore LH secretion. These data signify that caloric deprivation represseshypothalamic GnRH outflow to pituitary gonadotrope cells, which retain responsiveness to GnRH stimulation.(Reprinted with permission from Aloi et al. 1997.)

unrestricted caloric ingestion (Bullen et al. 1985). Ineach case, menstrual cycles became normal within 6 months of completing the study.

Observational studies

Cross-sectional appraisal of hypothalamo–pituitary–ovarian hormones in collegiate long-distance run-ners has revealed a subset of subjects with irregularmenstrual cyclicity due presumptively to reducedhypothalamic GnRH release (Rogol et al. 1983;Veldhuis et al. 1985; MacConnie et al. 1986) (Fig. 6.2).Blunted GnRH secretion in this setting is inferredfrom decreased LH pulse frequency in the pre-sence of normal or accentuated pituitary LH release

induced by the injection of synthetic GnRH peptide(Fig. 6.3). Precisely why LH pulsatility is disruptedin some but not all long-distance runners is notknown. Epidemiological correlations indicate thatone or more apparent reproductive risk factorsoften prevail (see below).

Table 6.1 highlights several factors that mayincrease the risk of developing irregular menstrualcycles in strenuously training young women.Among diverse considerations, deficient caloricintake in relation to any given level of energy expend-iture is an established risk for oligomenorrhea in prospectively randomized analyses (vide infra)(Bullen et al. 1985; Loucks 2003). Albeit not definit-ive, several other inferred clinical associations are

Page 89: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the reproductive axis 73

helpful in adumbrating possible avenues for furtherinvestigation (Veldhuis et al. 1998).

Insufficient total caloric intake, anorexia ner-vosa, bulimia, binge eating, surreptitious vomitingand excessive use of purgatives all activate thestress-adaptive hypothalamo–pituitary–adrenal axis.Complex central-neural responses to metabolicstress inhibit LH pulsatility by repressing hypotha-lamic GnRH release, thereby reducing feedforward

stimulation of LH secretion by the anterior pituitarygland (Bergendahl et al. 1996, 2000; Loucks 2003).For example, short-term fasting suppresses LHpulses consistently in healthy adults. A study inyoung men showed that the 50% fall in LH andtestosterone concentrations induced by fasting wasprevented by injecting pulses of synthetic GnRHevery 90 min intravenously (Aloi et al. 1997) (Fig.6.4). The outcome of this intervention demonstrates

GnRH2.5 µg

0 5 10Sample number

15 20 25 30

GnRH5.0 µg

GnRH10.0 µg

GnRH25.0 µg

ControlsRunners

60

50

40

30

20

10

Seru

m L

H (

mIU

·mL–1

)

Fig. 6.3 Accentuation of low-dose gonadotropin-releasing hormone (GnRH) -stimulated luteinizing hormone (LH)release in cross-sectionally recruited female collegiate long-distance runners with irregular menstrual cycles (see Fig. 6.2).Synthetic GnRH peptide was injected intravenously in escalating doses at the indicated times. LH concentrations weremonitored by sampling blood from the forearm every 20 min. Greater LH secretion stimulated by low doses of GnRH inwomen athletes than controls indicates that the pituitary gland is capable of producing LH in response to an adequate(hypothalamic) GnRH stimulus. Clinically inferred risk factors associated with menstrual dysfunction in such individualsare highlighted in Table 6.1. (Adapted with permission from Veldhuis et al. 1985.)

Table 6.1 Factors associated with increased prevalence of menstrual irregularity in athletes studied cross-sectionallyand/or by self-referral to a physician.

Apparent risk factor Putative mechanism

Self-imposed dieting, heavy laxative use, anorexia Metabolic and stress-adaptive reduction in hypothalamic or bulimia GnRH release

Anabolic steroids, testosterone and progestins Direct feedback suppression of LH and GnRH secretionHigh doses of injected human growth hormone Prolactin-like effect to limit brain GnRH secretionPsychosocial stress Central nervous-system inhibition of GnRH outflowPersonal or family history of reproductive disorder Unrelated primary disease; self-selection for study

GnRH, gonadotropin-releasing hormone; LH, luteinizing hormone.

Page 90: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

74 chapter 6

that caloric deficiency limits brain outflow of GnRH,but does block the pituitary gland.

Abuse (non-therapeutic administration) of ana-bolic steroids, testosterone, synthetic progestins andhuman growth hormone (which exerts a prolactin-like inhibitory effect on GnRH secretion) can causeoligo- or amenorrhea (fewer than normal or no men-strual cycles, respectively) and oligo- or azoos-permia (reduced or undetectable sperm counts).Erythropoietin injections are not known to do so.

A high degree of perceived psychosocial stress,whether originating from or independently of thetraining and competition environment, may alsodisrupt menstrual function (Evans et al. 1992;Veldhuis et al. 1998). In addition, some athletesbegin intensive training with pre-existing (unrecog-nized) hormonal abnormalities and/or a stronggenetic potential for endocrine disease. Both factorsmay be unmasked by strenuous athletic activity ordetected independently of the training experience.

15

10

5

0

(a) 0800 02001400 2000 0800

12.5

10.0

7.5

5.0

2.5

0.0

(b) 0800 02001400 2000 0800

8

6

4

2

0(c) 0800 02001400 2000 0800

40

30

20

10

0(d)

Clock time

0800 02001400 2000 0800

Seru

m L

H (

mIU

·mL–1

)

Fig. 6.4 Illustrative patterns of pulsatile luteinizing hormone (LH) release monitored every 20 min over 24 h in four young women identified cross-sectionally. Subjects were intercollegiate long-distance runners who reported irregularmenstrual cycles. Some participants had a subnormal frequency (daily number) of LH pulses. The causal relationship, if any, to strenuous physical training is unknown. As discussed in the text, an independent longitudinal investigationdemonstrated normal LH pulsatility before and after 1 year of endurance running in young women, each of whom ran at least 500 miles (800 km) (Rogol et al. 1992). (Reprinted with permission from Veldhuis et al. 1985.)

Page 91: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the reproductive axis 75

Unresolved issues

A significant and unresolved clinical issue is howtwo or more factors that individually heighten therisk of altered menstrual function interact adverselyin the setting of vigorous physical training. Thisquestion is particularly difficult to address, becausepossible combinatorial factors are nearly illimitable.For example, what would be the overall effect on menstrual function of strenuous training in awoman with a family but not a personal history ofpolycystic ovarian syndrome (present in 5.0–8.5%of otherwise healthy young women of diverse ethnicities), occasional prior menstrual irregularityand minimal weight loss during the exercise pro-gram? And how is the foregoing aggregate likeli-hood further influenced by the choice of a particularsport; for example, competitive fencing, volleyball,pole vaulting, the 500-m run or 100-m free-styleswimming. Observational inferences suggest thatmenstrual dysfunction is less common among par-ticipants in cycling and swimming than other athletic events (Evans et al. 1992). And under whatcircumstances are risk factors that are inferable inwomen applicable to men? There are no explicitdata at present to address such multifactorialqueries. In fact, bias of ascertainment, self-referral tophysicians, exercise modality (e.g. greater or lessercooling of core body temperature), perceived stress,probability of hormone abuse, weight loss or main-tenance, age of training onset and other unknownconfounding factors vitiate definitive interpretationof cross-sectional data.

In relation to the medical care of competitive ath-letes, doctors should be cognizant that physicalexertion alone is an unlikely proximate cause of clinically significant endocrine changes. Thus,physician-directed review of dietary habits, weightloss, caloric intake, medical records, family history,systemic symptoms, physical signs and screeninglaboratory tests can protect the athlete’s personalhealth. Thorough assessment is necessary to excludeor identify remediable associated illness.

Whether strenuous physical training affectssperm function or female fecundity (ability to con-ceive) or fertility (carrying a healthy infant to full-term delivery) has not been established rigorously.

Relevant investigations would comprise longitud-inal, unbiased monitoring of FSH, inhibin and act-ivin subunits, follistatin, spermatogenesis, Graafian follicle maturation, ovulation, luteinization, andlifetable analyses of fecundity, fertility and livebirths inter alia. Anecdotal clinical observations pre-dict that changes in fecundity or fertility would besubtle rather than clinically significant.

The impact, if any, of physical exertion on thepostpartum state is unknown. Illustrative unex-plored issues include the time required for recoveryof menstrual cyclicity after pregnancy; lactationalefficiency, milk volume and nutritional content forthe infant; and infant–maternal bonding.

Greater clinical understanding is required of therelatively neglected domain of sexual libido, poten-tia and satisfaction in athletes pursuing elite phys-ical training. And, medical assurance is needed thatall athletes have full access to instructional guidesconcerning risks of sexually transmissible diseaseand communicable respiratory and gastrointestinalillness. Both necessities arise from the wide spec-trum of sociocultural, nutritional and geographicvenues engaged in international competition.

Summary

Strenuous physical training in the healthy youngadult (albeit not necessarily in children) does notdisrupt reproductive function significantly. Thisconclusion does not apply when training is accom-panied by deficient caloric intake, weight loss, aber-rant eating habits, concomitant disease, high geneticrisk of a primary reproductive disorder, undue psy-chosocial stress and/or abuse of alcohol, drugs orhormonal agents. Additional clinical research isneeded in behalf of the athletic and medical commun-ities to clarify more subtle questions related to theimpact of strenuous physical exertion on fecundity,fertility, postpartum health and sexual function.

Acknowledgments

This work was supported in part by the NationalInstitutes of Health and the National Center forResearch Resources (Bethesda, Maryland, USA) viaRR00585, AG23133 and DK60717.

Page 92: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

76 chapter 6

References

Aloi, J.A., Bergendahl, M., Iranmanesh, A.& Veldhuis, J.D. (1997) Pulsatileintravenous gonadotropin-releasinghormone administration averts fasting-induced hypogonadotropism andhypoandrogenemia in healthy, normal-weight men. Journal of ClinicalEndocrinology and Metabolism 82,1543–1548.

Bergendahl, M., Vance, M.L., Iranmanesh,A., Thorner, M.O. & Veldhuis, J.D. (1996)Fasting as a metabolic stress paradigmselectively amplifies cortisol secretoryburst mass and delays the time ofmaximal nyctohemeral cortisolconcentrations in healthy men. Journal ofClinical Endocrinology and Metabolism81(2), 692–699.

Bergendahl, M., Iranmanesh, A., Pastor, C.,Evans, W.S. & Veldhuis, J.D. (2000)Homeostatic joint amplification ofpulsatile and 24-hour rhythmic cortisolsecretion by fasting stress in midlutealphase women: concurrent disruption of cortisol–growth hormone, cortisol–luteinizing hormone, and cortisol–leptin synchrony. Journal of ClinicalEndocrinology and Metabolism 85(11),4028–4035.

Bullen, B.A., Skrinar, G.S., Beitins, I.Z. et al.(1985) Induction of menstrual disordersby strenuous exercise in untrained

year of endurance training. Journal ofApplied Physiology 72(4), 1571–1580.

Urban, R.J., Evans, W.S., Rogol, A.D. et al.(1988) Contemporary aspects of discretepeak detection algorithms. I. Theparadigm of the luteinizing hormonepulse signal in men. Endocrine Reviews 9,3–37.

Veldhuis, J.D. (1996) Neuroendocrinemechanisms mediating awakening ofthe gonadotropic axis in puberty.Pediatric Nephrology 10, 304–317.

Veldhuis, J.D. (1999) Male hypothalamic–pituitary–gonadal axis. In: ReproductiveEndocrinology (Yen, S.S.C., Jaffe, R.B. &Barbieri, R.L., eds.). W.B. Saunders Co.,Philadelphia: 622–631.

Veldhuis, J.D., Evans, W.S., Demers, L.M.et al. (1985) Altered neuroendocrineregulation of gonadotropin secretion inwomen distance runners. Journal ofClinical Endocrinology and Metabolism 61,557–563.

Veldhuis, J.D., Yoshida, K. & Iranmanesh,A. (1998) The effect of mental andmetabolic stress on the femalereproductive system and femalereproductive hormones. In: Handbook ofStress Medicine: an Organ SystemApproach (Hubbard, J. & Workman, E.A.,eds.). CRC Press, Boca Raton, FL:115–140.

women. New England Journal of Medicine312(21), 1349–1353.

Evans, W.S., Sollenberger, M.J., Booth, Jr.,R.A. et al. (1992) Contemporary aspectsof discrete peak detection algorithms. II.The paradigm of the luteinizinghormone pulse signal in women.Endocrine Reviews 13(1), 81–104.

Giustina, A. & Veldhuis, J.D. (1998)Pathophysiology of the neuroregulationof growth hormone secretion inexperimental animals and the human.Endocrine Reviews 19(6), 717–797.

Loucks, A.B. (2003) Energy availability, not body fatness, regulates reproductivefunction in women. Exercise and SportsSciences Reviews 31(3), 144–148.

MacConnie, S.E., Barkan, A.L., Lampman,R.M., Schork, M.A. & Beitins, I.Z. (1986)Decreased hypothalamic gonadotropin-releasing hormone secretion in malemarathon runners. New England Journalof Medicine 315, 411–417.

Rogol, A.D., Veldhuis, J.D., Williams, F.T. & Johnson, M.L. (1983) Pulsatilesecretion of gonadotropins and prolactinin male marathon runners: relation tothe endogenous opiate system. Journal ofAndrology 5, 21–27.

Rogol, A.D., Weltman, A., Weltman, J.Y. et al. (1992) Durability of the reproductiveaxis in eumenorrheic women during 1

Page 93: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

77

Introduction

Exercise stimulates release of human growth hor-mone (GH) from the anterior pituitary gland and elev-ated concentrations of circulating GH ultimatelyhelp contribute to the exercise-induced increases inmuscle hypertrophy and fat breakdown as well asother physiological responses. A review of the exer-cise/GH literature permits the generalization that itis largely the duration and intensity of the exercisebout that regulates plasma concentrations of circu-lating GH.

It seems far less certain, however, that many arefully aware of the complexities underlying the GHproduction ‘system’ in the pituitary gland itself.Issues underlying these complexities often appearas studies published in specialty journals in whichthe major focus of the effort is not that of exercise-induced release of GH. This situation has started tochange.

For the reader primarily interested in exercisephysiology/medicine, the primary purpose of thischapter is to bring the issue of GH system complex-ity into sharper focus. What do we mean by ‘systemcomplexity’? Elements of that complexity reside insignals delivered to the pituitary from other parts ofthe body, for example muscles. Still other elementsreside in the GH producing cells themselves. Thefinal product of that complexity is the well-knowntextbook form of GH molecule, as well as variants ofthat molecule, that circulate in human blood. Wecontend that it is this ‘army of molecules’ that parti-cipates in bringing about the physiological responsesthat have been ascribed to GH for many years.

In this chapter we consider the biochemistry andphysiological activities of the GH molecule and itsvariant forms. We examine the literature that showsthat GH variants in the circulation change after exer-cise. We also briefly review aspects of the cellularbiology of the pituitary with the hope that theseaspects help offer a fundamental basis for appreciat-ing how production of GH variants may relate to theissue of what could happen after bouts of aerobic/resistance exercise. And finally we offer data thatstrongly suggest the existence of a novel feedbackloop from the muscle to the pituitary. The authorsbelieve that this newly discovered feedback loopmight help explain one mechanism(s) underlyingexercise-induced release of GH.

Measurement of human growthhormone

In today’s world, plasma concentrations of cir-culating GH are almost always measured usingimmunoassay procedures. However, other detec-tion systems exist and the method chosen to measure GH in the blood is important. Before theintroduction of GH immunoassays investigatorsusually relied on biological assays that oftenrequired the use of rats; certain of these bioassayscontinue to be used in the authors’ laboratories(Roth et al. 1963; Hunter & Greenwood 1964).Because they often yield interesting data that may attimes conflict with those obtained by immunoassay,we believe a review of such bioassay techniques iswarranted.

Chapter 7

Growth Hormone Variants and Human Exercise

WESLEY C. HYMER, RICHARD E. GRINDELAND, BRADLEY C. NINDLAND WILLIAM J. KRAEMER

Page 94: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

78 chapter 7

Growth hormone bioassays:perspectives from the older literature

Tests that were developed over 50 years ago re-flected the growing awareness at that time of theanabolic, lipolytic and diabetogenic actions of GH.A 1962 review (Papkoff & Li 1962) nicely sum-marized the essential details of some of these testsavailable at that time.

In terms of body weight tests the following gener-alizations apply: large numbers of rats (10/doselevel) are required; daily injections can be subcu-taneous or intraperitoneal; either intact or hypophy-sectomized rats are used; weight gain tests arerelatively insensitive (50 µg·day–1 in the intact mat-ure female rat, 10 µg·day–1 in the immature hypoxrat); indices of precision (calculated by dividing thestandard deviation of the responses by the slope ofthe line) are > 0.2; and finally other hormones con-taminating a native GH preparation (e.g. thyroxine)may synergize with GH resulting in augmentedweight gain. In spite of these disadvantages, the traditional rat weight gain bioassay, that measuresthe weight of hypox rats after subcutaneous admin-istration of GH for 10 days, is still required by regulators in the USA for individuals assessing thebioidentity and potency of GH preparations pro-duced by recombinant technologies.

In terms of the rat tibial line GH bioassay pro-posed by Greenspan et al. (1949), a major advantageover the weight gain bioassay was the markedincrease in assay sensitivity (a response can bedetected with a total dose of 5 µg administered overa 4-day period). This assay quantifies the width ofthe uncalcified epiphyseal cartilage plate delineatedfrom the silver nitrate stained calcified portions ofthe plate (Fig. 7.1). This test has been used by theauthors of this chapter in many studies, includingthose investigating effects of exercise/bed rest oncirculating GH.

The list of biological effects of human GHincreases steadily. As pointed out by Strasburger(1994), it has been known for a long time that GH isanabolic protein that promotes longitudinal bonegrowth. It is also lactogenic, has both insulin agon-istic and antagonistic properties, is lipolytic, stimu-lates ornithine decarboxylase in the liver, promotes

Page 95: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 79

sodium and water retention, and modulates immunesystem function among others.

The IM-9 lymphocyte cell line and the 3T3-F422Aadipocyte assays are more recent, noteworthyexamples of in vitro cell based biological assays. Toour knowledge they have not been used to evaluateGH activities of plasma after exercise. Space limita-tions prohibit their consideration in this chapter.

Growth hormone bioassays: new perspectives

In an excellent study published by Roswall et al.(1996), careful comparisons are offered between twonew GH bioassays developed in their laboratoriesand the hypox rat weight gain bioassay described insection earlier. In order to fully appreciate the basisfor these new assays it is necessary to consider: (a)details of the structure of GH molecule produced by recombinant technology (recombinant humangrowth hormone, rhGH); (b) its structural variantsand degraded forms; and (c) molecular interactionsbetween these well-defined forms and the humangrowth hormone (hGH) receptor.

The primary structure of recombinanthuman growth hormone and itsassociated molecular landmarks

The linear sequence of the 191 amino acid (22 kDa)form of rhGH is shown in Fig. 7.2. This form, ofcourse, is identical to the native 22 kDa GHmolecule that is synthesized in the pituitary glandand released into the bloodstream on physiological

demand. A distinguishing structural feature is theposition of the cystine residues responsible for for-mation of the large internal disulfide loop and the smaller loop at the c-terminus. Also shown inFig. 7.2 is an enzymatic cleavage site betweenresidues threonine-142 and tyrosine 143; a cleavagethat results in a two-chain structure held together by these disulfide bridges. Generation of this two-chain form may result from the action of a mem-brane bound protease during secretion from thepituitary gland. On long-term storage of GH in solu-tion, deamidation of asparagine residues 149 and152, as well as loss of the first two residues at the N-terminus, can occur.

Many structural variants of the GH molecule arepresent in biological samples. In further studiesRoswall et al. (1996) generated two of these natur-ally occurring variants using rhGH as their start-ing preparation. One was the covalent dimer ofmethionyl GH; the other was the 20 kDa trans-criptional variant produced by deletion of residues32–46. These variants were used in studies laterdescribed.

How rGH molecules interact with membranebound tissue GH receptors (GHRs) is obviouslyimportant for a more complete understanding of the importance and physiological consequences of exercise-induced elevations in circulating GH.Studies by Cunningham and his colleagues ~ 15years ago not only established the complete aminoacid sequence of membrane bound GHR, but alsoshowed that the extracellular domain was essen-tially identical to the glycosylated form of the recep-tor isolated from human serum (Cunningham &Wells 1989; Cunningham et al. 1991). These invest-igators learned that one molecule of 22 kDa GH complexed with two molecules of the extracellulardomain of GHR. At low GH concentrations receptorbinding occurs sequentially at two distinct sites onthe hormone molecule. Figure 7.3 shows detailsunderlying receptor dimerization that will lead tosignal transduction in GH responsive tissues.

Understanding the basis of this detailed molecularphysiology enabled Roswall et al. (1996) to developtwo different types of GH bioassays. One, termedhigh performance receptor binding chromatography(HPRBC), compares the ability of a test sample of

Fig. 7.1 (opposite) Rat epiphyseal cartilage plates preparedfrom hypophysectomized female rats after subcutaneousinjection of saline or growth hormone (GH) standard for 4days prior to sacrifice. The total dose of GH administeredis shown in the lower left corner of the panels. In this assaycartilage plates are stained with silver nitrate and platewidths measured with an ocular micrometer. Tenmeasurements are taken across the plate and averaged.Plates from control animals typically average 150 µm and those from GH injected rats increase to > 200 µm indose-dependant fashion. The assay is usually done indouble-blind fashion. Photographs kindly provided byDr. Scott Gordon.

Page 96: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

80 chapter 7

GH with that of a rhGH standard to form a stable 2 : 1 receptor/rGH complex with soluble GHR. Non-denaturing size exclusion chromatography is usedto analyze the resulting complex. Strasburger andcolleagues have recently developed the immuno-functional enzyme linked immunoassay (IFA) thatis based on these earlier findings of Cunninghamand his colleagues (Strasburger et al. 1996). This assayuses a GH monoclonal antibody and a biotinylated

rGH binding protein (BP) to assess functional activ-ity of the GH-containing preparation. The IFA hasbeen used by to measure circulating levels of GHafter exercise (Nindl et al. 2000)

In the second assay described by Roswall et al.(1996), termed the cell proliferation assay (CP), cellsfrom a mouse myeloid leukemia cell line (FDC for ~ 20 h prior to -P1) are transfected with full lengthreceptor and subsequently incubated with test

Denotes tryptic cleavage site

Denotes theoretical peptide number

des-Phe1 Pro2–hGH cleavage site

2-chain cleavage site

Oxidized methionine

Deamidated asparagine

Fig. 7.2 The linear sequence of recombinant human growth hormone (rhGH). The diagram indicates: (a) tryptic cleavagesites; (b) theoretical peptide number (T1–T21); (c) the two-chain enzymatic cleavage site; (d) methionine residuessusceptible to oxidation; and (e) asparagines residues susceptible to deamidation. Residues 32–46, marked in black, aredeleted in the 20 kDa splice variant of GH. (Adapted from Roswall et al. 1996.)

Page 97: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 81

samples measuring 3H-thymidine uptake into DNAas a measure of biological activity. A similar strategywas used by Wada et al. (1998) to measure GH activity of certain variants using Ba/F3-hGHR cells.These cell based assays measure a response (i.e.DNA synthesis) that is several steps distal to recep-tor dimerization. Roswall et al. (1996) suggest thatthis fact puts the assay: ‘several steps closer to the in vivo biological response’ (p. 36).

Assay comparisons

A comparison between the activities of genetic andchemical rGH variants assessed by the rat weightgain, HPRBC and CP assays are informative andimportant to consider before assessing similar

parameters in human plasma after exercise. Data inTable 7.1, reproduced from the Roswall et al. (1996),show full activity and good correspondence betweenresults in some samples (e.g. deamidated rGH andoxidized rGH); poor activity with preparations ofdimer or trypsin treated rhGH; and ‘super-potency’in the rat bioassay when the two-chain rhGH vari-ant is tested. As indicated by Roswall et al. (1996)and others, increased biological activity two-chainGH has been reported previously.

Even though the cell assays described by Roswallet al. (1996) and Wada et al. (1998) have yet to be usedfor studies using blood samples collected before andafter exercise stress, it seems quite likely that theywill in the near term become important for under-standing GH activity.

hGH

Receptor

Sequential binding*Site 1

Sequential binding*Site 1*Site 2

Signaltransduction

Fig. 7.3 Idealized model of a 22 kDa human growth hormone (hGH) molecule in plasma interacting with receptors on theplasma membrane of a target cell. Proper signal transduction requires receptor dimerization via two binding sites on thehormone molecule.

Sample description Rat bioassay HPRBC assay CP assay

Deamidated rhGH 0.88 (n = 1) 1.01 (n = 3) 0.86Two-chain rhGH 1.78 (n = 2) 0.75 (n = 2) 0.77Aspartic acid mutant rhGH N149D 0.80 (n = 1) 1.00 (n = 4) 0.87Aspartic acid mutant rhGH N152D 1.22 (n = 1) 0.95 (n = 2) a

Aspartic acid mutant rhGH 0.85 (n = 1) 0.96 (n = 4) 0.93N149D + N152D

Des-Phe1Pro2-rhGH 0.46 (n = 1) 0.60 (n = 4) 0.94Oxidized rhGH 0.79 (n = 4) 0.98 (n = 2) 0.77Met-hGH covalent dimer 0.10 (n = 1) 0.07 (n = 5) 0.36Aged rhGH 0.73 (n = 1) 0.96 (n = 3) a

Trypsin-treated rhGH 0.47 (n = 1) 0.30 (n = 1) a

See Roswall et al. (1996) for a discussion of the principles underlying these data.Note that the two-chain variant, relative to the 22 kDa form, is highly potent inthe rat weight gain assay and less so in the other in vitro biological assays.

Table 7.1 Biological activities ofgenetic and chemical variants ofrecombinant human growthhormone (rhGH) by the rat weightgain assay; the high performancereceptor binding chromatography(HPRBC) assay and the cellproliferation (CP) assay. (Data taken from Roswall et al. 1996.)

Page 98: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

82 chapter 7

The wide array of growth hormoneforms in the blood

It has been suggested by Baumann (1991b) thatthere may be as many as 100 different forms of GHpresent in the human circulation. The concept thatnumerous molecular forms of the hormone arisefrom post-transcriptional/translational modifica-tion of the single pituitary GH-N gene is certainlynot new. Pioneering work from the laboratories ofLewis, Sinha, Kostyo and Baumann to name but a few, provide the foundation for the informa-tion offered in Table 7.2 that is taken from a 1991

Baumann paper (Baumann 1991b). It shows the estimated proportions of GH variant forms in theplasma 15 min after a secretory episode. Many ofthese studies were done before techniques of recom-binant technology became widely available. Notsurprisingly, this type of information therefore wasobtained using traditional biochemical procedures.

The literature is rich with observations concern-ing the biochemical nature of GH variants. A briefbut non-comprehensive review of this literature isimportant for the innate understanding of the het-erogeneity and complexity of the GH mechanismspotentially involved with exercise responses and

Proportion of total GH GH form (%)

Monomeric22 kDa total 43.0

22 kDa free 21.022 kDa in high affinity complex 20.022 kDa in low affinity complex 2.0

20 kDa total 8.020 kDa free 5.520 kDa bound in high affinity complex 0.520 kDa bound in low affinity complex 2.0

Acidic GH (desamido- and acyl-GH) total 5.0Acidic GH bound fractions Unknown

Dimeric22 kDa non-covalent dimer total 14.022 kDa disulfide dimer total 6.0

22 kDa dimers bound fraction Unknown20 kDa non-covalent dimer total 5.020 kDa disulfide dimer total 2.0

20 kDa dimers bound fraction UnknownAcidic GH non-covalent dimer total 1.5Acidic GH disulfide dimer total 0.5

Acidic dimers bound fractions Unknown

Trimeric to pentameric22 kDa non-covalent oligomers total 7.022 kDa disulfide oligomers total 3.0

22 kDa oligomers bound fraction Unknown20 kDa non-covalent oligomers total 1.020 kDa disulfide oligomers total 0.5

20 kDa oligomers bound fraction UnknownAcidic GH oligomers (non-covalent and S–S) total 1.0

Acidic GH oligomers bound fraction UnknownNon-S–S-linked covalent oligomers total 1.0

Fragments16 kDa, 12 kDa, and 30 kDa immunoreactive fragments Variable

Table 7.2 Estimated proportions ofgrowth hormone (GH) variant formsin human plasma 15 min aftersecretion. (From Baumman 1991b.)

Page 99: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 83

adaptations. The fact that plasma GH immunoreact-ivity consists of several molecular weight speciesthat can be separated by size exclusion column chro-matography has been known for > 30 years. In thepast it was useful to categorize the three main isomers (variants) of GH as little, big or big–big; adesignation based upon their elution positions fromthe column. The physical nature of these size vari-ants is not nearly as well defined as those in studiesusing GH made by recombinant means. Neverthe-less, the careful studies by the Baumann (Baumann1991a, 1991b, 1999; Baumann et al. 1994) and Lewisgroups (Lewis et al. 2000), among others, that haveattempted to characterize the big and big–big variants, permit the conclusion that these variantsrepresent an oligomeric series. Similar oligomers inextracts of human pituitary tissue further supportthis view. Most investigators therefore believe thataggregation states up to at least pentameric hGHexist and the distinction of big and big–big GH is arbitrary. The authors prefer to categorize theoligomers in terms of their apparent molecular massbased on elution profiles following sephadex chro-matography. In addition to these oligomeric sizevariants charge variants of the GH molecule areknown. These are thought to be reflected in acety-lated, deamidated or cleaved GH (see Fig. 7.2).

The careful study by Stolar et al. (1984) alsoshowed that a majority of the big and big–big GHvariants converts to little hGH (22 kDa) duringextraction and storage (e.g exposure to 4 mol potas-sium thiocynate [KSCN] and two freeze–thawcycles result in ~ 70% conversion to little GH). Thevariants surviving this harsh treatment migrate asdistinct species with apparent molecular weights of45, 62, 80 and 110 kDa. These latter forms covertquantitatively (and mainly) to little GH after sulfhy-dryl reduction. Acidic GH comprises a smaller com-ponent. The 20 kDa variant may tend to dimerizepreferentially.

What is known about the biological activity ofthese oligomeric species? In general, the big forms(dimer) are thought to have reduced activity inradioreceptor and rodent based assays. However, inthe IFA, Strasburger et al. (1996) report that dimers,on a molar basis, have slightly higher reactivity inthe IFA (110%) relative to the 22 kDa monomer.

A recent review by the Lewis and Sinha groupsummarizes the properties of five variant forms ofGH (Lewis et al. 2000). Two of these are the short andlarger peptides generated from proteolytic cleavagebetween residues 43 and 44 of the GH molecule (seeFig. 7.2). Their evidence favors the concept that the short peptide (GH [1–43]) potentiates the physi-ological effects of insulin; the larger peptide (GH[44–191]) has anti-insulin properties. In fact theseauthors indicate that: ‘. . . we believe that this (largerpeptide) is the long sought after diabetogenic sub-stance of the pituitary gland’ (p. 58).

A ~ 3 kDa peptide, isolated from both humanpost-mortem pituitary tissue and human plasma, isactive in the rat tibial line bioassay (Hymer et al.2000). This peptide is not a fragment of the GHmolecule. The relationship of this finding to variantforms described by Baumann is unclear (Baumann1999). Limited amino acid sequence data, includingresidues 9–25 in the middle of the peptide, indicatethat this peptide is not a breakdown product of theGH molecule. Most interesting is the finding thatmany of these residues are non-polar and bear strik-ing similarity to the c peptide that is contained in theproinsulin molecule. Similar to the c peptide, thishuman pituitary tibial peptide, obviously has bio-logical activity. Unpublished data from one of ourlaboratories (R.G.) shows that the rat pituitary alsocontains a small peptide that is active in the tibialbioassay.

Variant forms of growth hormone: an emerging data base

In spite of the fact that it has been known for manyyears that exercise is a potent stimulator of circulat-ing GH, it is only recently that the topic of possibleexercise-induced generation of circulating GH vari-ant forms been addressed at all (Nindl et al. 2003). Inthis section we consider some of our prior informa-tion in the context of the background given previ-ously. To summarize and analyze this informationin a logical way requires an appreciation of the following types of variables in the analysis: humansubject choice; exercise type (intensity/duration);GH assay type; methodology used to isolate the vari-ant forms; and special treatments of blood samples.

Page 100: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

84 chapter 7

Table 7.3 provides a summary by subcategorizingstudies (only human) according to our arbitraryrequirement that the blood sample has been studiedby at least two procedures for the purposes of fur-ther understanding exercise-induced rises in circu-lating GH in terms of GH molecular heterogeneity.Only the Hymer study (Hymer et al. 2001) used frac-tionated plasma to measure GH variants; all the restin Table 7.3 used only neat plasma.

Data from studies to date thus far show that exercise can modify either the activity or molecularcharacter of GH in the circulation. Wallace et al.(2001) utilized seven different assays to measureGH in 17 aerobically trained men before and after 20 min of cycle ergometry at 80% Vo2max to furthercharacterize the response of GH molecular isoformsto exercise. Serum was assayed with antibodiesspecific for total, pituitary, 22 kDa, recombinant,non-22 kDa, 20 kDa and immunofunctional (IF)GH. Salient findings from this study were: (a) allforms of GH increased during and at the end ofexercise; (b) 22 kDa GH was the predominant isoform (73%) at the cessation of exercise; (c) theratios of non-22 kDa/total GH and 20 kDa/total

GH increased and those of recombinant/pituitaryGH decreased. Wallace et al. (2001) attributed theincrease in non-22 kDa isoforms to slower disap-pearance rates of 20 kDa and perhaps non-22 kDaGH isoforms. Collectively, Wallace’s findingsdemonstrate that the proportion of GH isoformchanged across acute exercise and into recovery.Although the 22 kDa was the predominant isoformdetected at peak concentrations, isoforms of GHother than 22 kDa increased during the post-exercise period. These results suggest that the pro-portion of 20 kDa, 17 kDa and possibly other non-22 kDa isoforms (dimers, oligomers and GHbound to serum proteins) increase after exercise.The authors postulated that the increase in the pro-portion of isoforms other than 22 kDa after exercise may be attributed to differential pituitary isoformsecretion, the appearance of isoforms from non-pituitary sources, generation of fragments, dimersand oligomers in the circulation, and differences inclearance rates of the different isoforms. Theauthors also speculated that the biological conse-quences of their findings might potentially reside in enhanced diabetogenic effects of smaller GH

Table 7.3 Summary of recently published studies in which different types of growth hormone (GH) assays have beenused to measure concentrations of GH in human plasma after aerobic or resistance exercise.

GH GH immunoassay bioassay

Chemical Reference Exercise type/duration IRMA Poly Tibial IFA reduction

Rubin et al. 2003 Treadmill: 60, 75, 90, 100% Vo2max X X Xfor 10, 10, 5 & 2 min, respectively

Hymer et al. 2001 Squats: 6 sets of 10-RM X X X X XWallace et al. 2001 Cycle: 80% Vo2max for 20 min X XNindl et al. 2001 Resistance: high volume, multiset X X XBigbee et al. 2000 Treadmill: 27 m·min–1 for 15 min X XMcCall et al. 2000 Vibration: stimulus-muscle X X

afferents for 10 minNindl et al. 2000 Squats: 6 sets of 10-RM X XMcCall et al. 1997, 1999 Isometric plantar flexion: 30, 80, X X

100% MVC

Poly: assay using a polyclonal antiserum.Tibial: rat tibial line bioassay.Chemical reduction: those studies in which a reducing agent has been added to the plasma sample before assay.IFA, immunofunctional assay (as described by Strasburger et al. 1996); IRMA, immunoradiometric assay; RM, repetitionmaximum.

Page 101: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 85

isoforms, which may serve to prevent post-exercisehypoglycemia.

Extending on Wallace’s work (Wallace et al. 2001),we (Hymer, Kraemer and Nindl) next conducted astudy in which we fractionated human plasma in 35women before and after acute resistance exercise(six sets of 10-repetition maximum [RM] squats,separated by 2-min rest periods) using size exclu-sion chromatography into three size classes (Hymeret al. 2001). Fraction A contained molecules > 60 kDa(presumably oligomers and/or monomeric GHbound to receptor); fraction B contained molecules30–60 kDa (presumably homodimers and hetero-dimers); and fraction C contained GH molecules < 30 kDa (presumably a mixture of 22, 20, 16, 12, and5 kDa forms). All samples were then assayed usingthe Diagnostic Systems Laboratory (DSL) IFA, theNichols radioimmunometric assay (IRMA) and theNational Institutes of Diabetes and Digestive andKidney Diseases radioimmunoassay (NIDDK RIA).Additionally, we assayed all samples before andafter glutathione (GSH) treatment in order to deter-mine the effects of chemical reduction of disulfidelinked bonds. Recovered immunoreactivities were4–11% in fraction A, 22–45% in fraction B and44–72% in fraction C. Significant exercise-inducedincreases were observed for the lower molecularweight GH moieties (30–60 kDa and < 30 kDa iso-forms), but not for the higher molecular weight GHmoieties (> 60 kDa). Another important finding wasthat chemical reduction of the post-exercise samplesincreased immunoassayable GH as measured by theNichols and NIDDK assays more than pre-exercisesamples, suggesting that exercise may specificallyincrease the release of disulfide-linked hormonemolecules and/or fragments. From these data, themost important effect of acute resistance exerciseappears to be on dimeric hormone. Because com-plexes of GH and BPs have longer half-lives thanfree GH, it is possible that dimeric GH might alsohave a longer half-life. Therefore, the net effect ofthe increase in GH isoforms within this molecularweight range would be to prolong the biologicalactivities of these forms after exercise.

In a study by Nindl et al. (2000) the first data com-paring the effects of exercise on IF versus immuno-reactive (IR) GH was presented. Comparisons were

made between the IF versus IR GH concentrationsin men and women before and after acute resistanceexercise (i.e. six sets of 10-RM squats separated by 2-min rest periods). IF GH concentrations weredetermined by an enzyme linked immunosorbentassay (ELISA) purchased from Diagnostics SystemsLaboratories (DSL, Webster, TX), which was basedon Strasburger’s work (Strasburger et al. 1996), andIR GH concentrations were determined by a mono-clonal IRMA purchased from Nichols (San JuanCapistrano, CA). In this study, both men andwomen demonstrated similar increases for IR (men:1.47 vs. 25.0 ng·mL–1; women: 4.0 vs. 25.4 ng·mL–1)and IF (men: 0.55 vs. 11.7 ng·mL–1; women: 1.94 vs.10.4 ng·mL–1) GH following exercise. However,post-exercise IF GH was significantly less than IRGH for both men and women. The ratio of IR/IFafter exercise was ~ 2 and similar for both men andwomen. The correlation between post-exercise IRand IF GH was r = 0.83. This study initially indic-ated that about half of the GH isoforms measured by the Nichols IRMA released after exercise did not possess intact sites 1 and 2 required for receptordimerization, thus suggesting biological inactivity.

A following study considered the fact that GH isreleased in an episodic, pulsatile manner. IF GH wasmeasured in 10 men who underwent two overnightblood draws with sampling every 10 min from 1700to 0600 h. The overnight serial sampling was per-formed in both a control and an acute heavy resist-ance exercise condition (Nindl et al. 2001). For the exercise condition, subjects performed a high-volume, multiset resistance exercise bout from 1500to 1700 h. IF GH was compared to the Nichols IRMAand NIDDK’s polyclonal RIA. The Pulsar peakdetection system was used to evaluate the pulsatil-ity profile characteristics of GH. Even though theresults from all three immunoassays were highlycorrelated (correlations ranged from 0.85 to 0.95),the Nichols IRMA again yielded higher mean GHconcentrations than did IF GH (3.98 vs. 1.83 ng·mL–1,respectively). The results from the Nichols IRMAalso yielded higher pulse amplitudes compared toIF GH (8.0 vs. 4.63 ng·mL–1, respectively).

The consistent finding in these studies (Nindl et al. 2000, 2001, 2003) was that IF GH measuredfrom the same sample was approximately one half

Page 102: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

86 chapter 7

that of the Nichols IRMA, one of the most widelyused GH assays in clinical practice in the USA. Sincethe IF assay purports only to measure biologicallyactive forms of GH (i.e. only those forms of GHcapable of inducing receptor dimerization are trans-lated to an assay signal), the additional GH isoformsdetected by the Nichols IRMA are likely fragmentswhose potential biological actions are not mediatedby the GHR. It has been reported that the GH frag-ment 44–191 is detectable in substantial levels inhuman serum and may even antagonize GH action(Rowlinson et al. 1996). Since this fragment lackspart of the N-terminus, it is unlikely to be detectedby the IF assay; however, the fragment could bedetected by the IRMA, depending on the targetedepitopes. Alternatively, the additional GH isoformsdetected by the Nichols IRMA could also be highmolecular weight variant forms of GH (Baumann1991a; Lewis et al. 2000).

Our findings conclusively show that at least someof the molecules released during secretory burstsare able to dimerize GHRs. In that sense, thesemolecules are biologically active. On the other hand,our data also demonstrated that GH isoforms arereleased, both in exercise and non-exercise con-ditions, that are not capable of initiating signaltransduction through the GHR. Based on the highcorrelations among the immunoassays and the similar detection of the number of peaks and inter-peak intervals, it appears that the immunoassaysreport qualitatively comparable pictures of the GHresponse. The quantitative differences among theimmunoassays have yet to be fully explained, butare likely due to the existence of various molecularisoforms. Other factors that contribute to the differ-ences in GH measurement could include the assayequilibrium conditions, buffer, tracer and standardused (Wood 2001).

It is important to consider the impact of BPs in theIFA (Strasburger et al. 1996; Nindl et al. 2001). TheIFA uses an rGHBP to bind site 1. One could inferthat a GH molecule already complexed to a GHBPwould not be detected in this assay system, as site 1would not be freely accessible. Also, a GH–BP com-plex might be configured such that site 2 is not ex-posed to the monoclonal antibody (mAb7B11). It hasbeen reported that the high-affinity GHBP inhibits

GH binding to receptors and in vitro bioactivity viacompetition for ligand (Strasburger et al. 1996). If itis true that GH complexed to BP is too large a mole-cule to traverse the capillary endothelium in orderto bind to cellular receptors, the lack of detection of the GH complexed in the IFA provides furthersupport for the functional selectivity of the IFA.

Rubin et al. (2003) also compared the Nichols IR versus the DSL IFA in six endurance-trained menduring intermittent treadmill running at progress-ively increasing intensities (60% Vo2 max for 10 min,75% for 10 min, 90% for 10 min and 100% for 2 min).Samples were assayed before and after the additionof glutathione (GSH; 10 mmol for 18 h at room temperature) in order to break disulfide bondsbetween possible oligomeric GH complexes. For theIRMA, GH was elevated after the 75% exerciseintensity and remained elevated through 30 min ofpost-exercise. After adding GSH, the IRMA indic-ated elevations in GH as early as 60% exercise intens-ity and remained elevated 45 min into recovery. At75%, the GSH assay run was higher than the non-GSH assay run. With the IFA, GH was elevated at60% in the non-GSH conditions, whereas the GSHassay run indicated elevations at 75%. Both GSH andnon-GSH conditions remained elevated through 30 min of recovery. These data indicate that theaddition of GSH to serum samples prior to assay viaan IRMA may break existing disulfide bonds aggreg-ated to GH molecules, thus altering the apparentassays signal to reveal total GH.

The complexity of the anterior pituitarycell system

The GH-producing cell system of the anterior pitu-itary gland is tremendously complex. How mightknowledge of that complexity help in unravelingmysteries and mechanisms underlying the relation-ships between exercise and GH variants? Humanpituitary tissue obtained after surgery or death isdifficult to obtain, so the rat pituitary gland has beenthe tissue of choice to study biology of the GH cell.Space limitations do not permit full review of thistopic. Instead, results from highly selected studiesare summarized here, in the format of a few sen-tences for each point considered.

Page 103: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 87

1 Human pituitary glands weigh ~ 350 mg; those of rats 8–12 mg. Human pituitary glands contain4–8 mg GH/gland; rats ~ 200 µg GH/gland. GHaccounts for ~ 80% of the total hormone in the gland.2 Quantitative extraction of GH requires homogen-ization at alkaline pH (9–10). Non-reducing sodiumdodecylsulfate polyacrylamide gel electrophoresis(SDS-PAGE) of pituitary extracts shows multipleGH forms with apparent molecular weights of14–88 kDa. After chemical reduction there is a fourto sixfold increase in the GH immunoreactivity inthose forms with mass > 50 kDa (Table 7.4). Highmolecular weight forms of GH are aggregates thatare linked by disulfide bridges resulting fromantiparallel alignment of cystine residues betweentwo GH monomers (Lewis et al. 1975). GH aggreg-ates are released from the pituitary and are presentin human plasma.3 Aggregates of GH are packaged in ~ 300 µ dia-meter secretion granules. These granules containbiologically active GH (Hymer & McShan 1963).According to Dannies (1999) ‘not a great deal’ isknown about molecular packaging of the hor-mone into granules and, ‘. . . how cells concentratehormones is a major unanswered question inendocrinology’ (p. 3). In some hormone-packagingcell systems there is good evidence that differentsecretory granule proteins are packaged in separategranules in the same cell. It is probable that hetero-geneity in GH packaging is directly related to theissue of GH variants and exercise.4 Subpopulations of rat GH containing granules are separable by continuous flow electrophoresis;

and GH containing granules isolated from the post-mortem human pituitary also yield subpopulationsas well. Preliminary evidence indicates that themore rapidly migrating particles are rich in tibialline bioactivity and relatively poor in IR GH.5 At least two types of rat pituitary GH cells can beroutinely separated by density gradient centrifuga-tion. GH that is rich in tibial line activity is preferen-tially released from the more dense cell; and thishormone is oligomeric (Farrington & Hymer 1990).6 Implantation of GH cells into the cerebral vent-ricles of hypophysectomized rats show that receip-ents gain weight and have longer tibial, femoral and pelvic bones (Weiss et al. 1978). Intraventricu-lar implantation of the denser GH cells not onlyincreases body weight, it also increases width of theepiphyseal plate and weight of the gastrocenimusmuscle (Grindeland & Hymer unpublished).

Collectively these findings support the contentionthat not only is there a molecular basis underlyingGH variants, but that there is an underlying cellularbiology basis as well.

Tibial line assay and exercise responses

The issue of the dichotomy between measurementsof GH concentrations made by tibial line bioassayand those made by immunoassay is important to consider in future experiments addressing therelationship between GH variants and exercise. It isimportant to consider GH variants in terms of thosemolecular forms which react with high affinity anti-sera to the 22 kDa ‘native’, textbook form of GH

GH (µg)

Apparent mol. wt. (kDa) –βME +βME Fold increase

< 22 4.7 + 0.9 6.1 + 0.7 1.322–57 1.8 + 0.2 3.2 + 0.5 1.857–77 2.1 + 0.2 8.1 + 0.4 3.977–100 1.2 + 0.2 4.7 + 0.5 3.9100–150 0.9 + 0.15 5.1 + 0.3 5.6< 150 0.9 + 0.3 4.0 + 0.4 4.4

Hormone contained in different regions of the gel were eluted and reduced withmercaptoethanol prior to GH using a polyclonal antibody. Recovery of GH fromthe gel averaged 103% (n = 3 experiments).

Table 7.4 The effect of chemicalreduction on concentrations of ratgrowth hormone (GH) variantscontained in alkaline extractsseparated on non-reducing sodiumdodecylsulfate polyacrylamide gelelectrophoresis (SDS-PAGE) ratpituitary prior to immunoassay.(From Farrington & Hymer 1990.)

Page 104: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

88 chapter 7

(immunoassayable GH; iGH) or to those form(s)that do not react with such antisera, but which elicitgrowth as revealed by the tibial line bioassay (bioas-sayable GH; bGH). It is possible that the pituitarysecretes variant forms of GH which have other func-tions (e.g. lipolytic activity), but literature on thispossibility is somewhat limited.

At the moment the tibial assay appears to be theassay of choice if one wishes to know the GH ‘status’of the subject. In spite of the fact that the assay islabor intensive, costly and lengthy it clearly offersinformation that is simply not obtainable any otherway. There is absolutely no question that today’sscientist or clinician is comfortable and familiarwith data that report plasma GH concentrationsmeasured by immunoassay. However, apparentconcentrations of bGH reported in the literature are often hundreds or even thousands of nanogramper milliliter! Why is this so? Because this bioassaymeasures the biological activity and not nanograms ofpurified hormone. In this context it is important torealize that purified GH from a number of differentspecies (human, bovine, murine), and their bGHs,yield parallel dose response curves in the tibialbioassay. Similar tibial growth curve responseselicited by these hormone preparations enable oneto express biological potency in terms of a standard22 kDa hormone preparation.

In any event it is important that, at the very least,some appreciation of the difficulty and importanceof establishing the most meaningful quantificationof plasma GH in a given physiological context of the subject is determined. Moreover any possibleskepticism regarding measurement of bGH by tibialassay may be alleviated by the following two exam-ples of the dichotomy issue between bGH and iGH.

Dichotomy between bioassayablegrowth hormone and immunoassayablegrowth hormone

The rat has played a major role in the evolution ofour current understanding of the iGH–bGH dicho-tomy. For example, many investigators report thatstimuli (e.g. cold exposure, hypoglycemia, exercise)that elicit increased plasma iGH concentrations inhumans have no effect on circulating iGH in the rat.

Investigations by one of us (R.G.) revealed that therat responded to these stimuli by secretion of GH inform(s) not recognized by antisera to the 22 kDa ratGH molecule (Ellis & Grindeland 1974). However,these circulating immunologically unreactive formspromoted significant growth of the assay rat (Ellis &Grindeland 1974).

These types of results permit the general, yet stillspeculative, conclusion that while there is no obviousrelationship between iGH and bGH in the rat, vari-ations in bioactive/immunoactive human GH concentrations in biological samples tend to changein the same direction. However, because titers ofhuman iGH and bGH are not directly proportionalwe believe that one should not use iGH measure-ments as an index of total circulating GH.

Almost coincidental with these early studies onrat and human bGH was the finding that plasmin, aprotease, was active on subprimate GH (rat, bovine)and either reduced or abolished the immunologicalactivity of the 22 kDa hormone (Ellis et al. 1968).However, this treatment yielded peptides with normal or even enhanced biological activity. Otherlaboratories have shown that human iGH, treatedwith human plasmin, does not appear to loseimmunological activity but to gain in biopotency(Singh et al. 1974; Lewis et al. 1975; Nguyen et al.1981). Clearly the immunological/biological ratio ofthe 22 kDa GH molecule can be significantly alteredafter enzyme treatment.

Human bed rest studies

Humans performing a moderate exercise (i.e. plan-tar flexion) lasting several minutes show a one totwofold increase in plasma bGH but no effect oniGH when done under the usual 1 G conditions.However, when subjected to absolute, head-downbed rest the identical exercise regimen does notevoke an increase in bGH secretion and has no effecton iGH release (McCall et al. 1997). Interestingly, afew days after bed rest the increased bGH secretionin response to exercise returns (Table 7.5).

What bearing might these data from have on GH variants and human exercise? In the exercisephysiology literature metabolic regulators are usu-ally invoked as a dominant mechanism by which

Page 105: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 89

increased muscle activity increases iGH secretion. Itis interesting to find that none of the commonlycited metabolic factors (e.g. lactate, plasma glucose)appear to explain reduced plasma concentrations of bGH. This obvious inconsistency led one of theauthors (R.G.) to ask the following question: ‘Isthere a neural mechanism that controls GH secre-tion?’ The answer seems to be yes; and it is relevantto exercise physiologists.

Muscle afferents regulate release ofbioassayable growth hormone in rats

Initial studies used animals in which nerves servinghind-limb muscles were severed. When the distalends are stimulated electrically for 15 min, a treat-ment that generates a pattern resembling that of arat walking at 1.5 miles per hour (2.4 km per hour)(Gosselink et al. 1998, 2000; McCall et al. 2000), the

stimulation has no effect on concentrations of eitherplasma or pituitary bGH or iGH.

On the other hand, if the proximal end of the severed nerve of a fast twitch muscle is stimulated,there is a massive (one to twofold) increase inplasma bGH within 5 min after stimulation! Sig-nificantly, this increase in plasma bGH is mirroredby a large decrease in pituitary gland concentrationsof bGH. However, there is no effect on iGH con-centrations in either plasma or the pituitary gland.Equally interesting is the finding that stimulation ofthe proximal end of the soleus nerve results inreduced plasma concentrations of bGH. This observa-tion implies specificity of muscle groups in thisafferent pathway.

These results are interesting from two points ofview. First, they argue that there is a neural controlcomponent to the pituitary GH system in additionto a metabolic component. Secondly, we wouldargue that these nerve stimulation studies provideinsight into the physiological significance of bGH. Ifone assumes that one of GH’s most critical functionsis assuring a constant glucose supply to the heartand brain, the results appear to make good sense.Massive GH release in response to high metabolicdemand (e.g. fasting, hypoglycemia or cold expos-ure) and release of GH in response to activation of aquiescent muscle of locomotion would both offermechanisms for increased carbohydrate uptake bytissues. These are protective mechanisms for theorganism.

Of course it is well established that human iGH issecreted in response to exercise, but this responsebegins only after 15 or 20 min of exercise. We pro-pose that during quiescence the soleus and otherpostural muscles, which are perhaps 80% active atrest, signal the pituitary via muscle afferents todecrease bGH secretion and thereby permit non-neural and non-cardiac tissues to use glucose. Asthe muscles of locomotion become active the mus-cles would signal the pituitary gland to secretebGH. The net effect would presumably be to inhibitutilization of glucose by active muscle and providean alternate energy supply by mobilizing fatty acidsfrom the fat depots.

In Fig. 7.4 we offer a model, adapted from ourpublished study (McCall et al. 2001), that closes a

Table 7.5 Plasma immunoassayable and bioassayablegrowth hormone concentrations. Pre- and post-exerciseduring bed rest (ng·mL–1). (Adapted from McCall et al.1997.)

Day Hormone Pre Post

Before bed rest–13/12 bGH 2146 0 3565*

iGH 5.3 4.7–8/7 bGH 2162 0 4161*

iGH 5.0 5.3

During bed rest2/3 bGH 2350 2203

iGH 5.1 5.88/9 bGH 2433 2105

iGH 4.8 5.213/14 bGH 2594 2085

iGH 4.7 5.2

After bed rest+2/3 bGH 1807 2379

iGH 2.0 4.5+10/11 bGH 1881 0 4160*

iGH 4.9 5.3

*p < 0.05 between pre- and post-exercise values. Growthhormone measured by immunoassay did not differ at any time or condition between pre- and post-exercisesamples. bGH, bioassayable growth hormone; iGH,immunoassayable growth hormone.

Page 106: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

90 chapter 7

muscle/neural feedback loop to the pituitary GHsystem. In Fig. 7.4 the neural inputs are received byhypothalamic neurons. On the other hand, it is con-ceivable that neural inputs might be received by theanterior pituitary gland directly. There are a fewreports in the literature that describe nerve inputs

to the pituitary. One of the more recent is that ofPaden et al. (1994) that describe ‘. . . a surprisinglyextensive innervation of the anterior lobe of thepituitary’ (p. 503). It is interesting that they are alsofrequently associated with blood vessels and do nothave the appearance of vasomotor fibers. Their dis-tribution is uneven and they appear to contact onlya subset of glandular cells (GH/adrenocorticotropichormone; ACTH).

Acute versus chronic resistance exerciseand bioassayable growth hormone

In a recent study we (Kraemer, Hymer and Nindl)evaluated the effects of an acute resistance exercisebout (i.e. six sets of 10-RM squats separated by 2 min of rest) on bGH before and after 6 months of periodized resistance training in young, healthywomen. Results from this study indicated that whileacute resistance exercise did not alter circulatingbGH, 6 months of chronic resistance training clearlypotentiated bGH concentrations (Fig. 7.5). Theseresults suggest that one of the benefits of chronicresistance training is to increase the biological activ-ity of circulating GH. This new finding potentiallyrepresents one of the mechanisms by which muscleand bone can benefit from resistance training.

Summary

The main points we have tried to emphasize in thischapter can be summarized in the following way.1 GH molecules are heterogeneous. In this chapter wedefine and consider GH heterogeneity in differentways. These include: (a) size and charge variants ofthe molecule(s) resulting from the single pituitaryGH gene; (b) activities of the molecule(s) in terms of the biological (in vivo) versus immunological (invitro) signals they generate; and (c) cells in the pitu-itary that produce and release GH molecules.2 Either aerobic or resistance exercise can result in differ-ential release of GH variants into the circulation. To datethere are only a handful of studies that address theissue of GH variants after human exercise. Thus farthey support the idea that release of oligomericforms of the 22 kDa GH monomer is increased afterexercise. The activities of circulating GH, measured

Gastrocnemius

Hypothalamus

Anterior pituitary

(Circulation)

Chronicunloading

BGHBone growthMuscle growth?Other functions?

GRF (+)SS (−)

Spinal cord

Tibialisanterior

Soleus

Low threshold afferent fibers, activated by:• Electrical stimulation• Exercise• Vibration

− +

Fig. 7.4 Model of proposed muscle afferent–pituitaryfeedback axis that is postulated to regulate release ofbioactive growth hormone (BGH) from the anteriorpituitary gland. BGH is defined as those form(s) of thehormone molecule that stimulate widening of theepiphyseal cartilage plate (see Fig. 7.1). This model issuggested by the studies of McCall et al. (2001) in whichstimulation of afferents from ankle flexors of the rat (e.g.tibialis anterior) or the entire posterior compartment ofankle extensors (e.g. soleus and gastrocnemius) stimulatesrelease of BGH but inhibits BGH when only afferents fromthe soleus muscle are stimulated. Chronic unloadinginhibits the exercise-induced increase of plasma BGH.Afferents may activate pituitary GH cells directly.Possible physiological functions of BGH, other thanstimulation of the tibial epiphyseal growth plate, have notbeen identified. GRF, growth hormone releasing factor; SS, somatostatin. (Adapted from McCall et al. 2001.)

Page 107: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 91

by biological and immunological assays, are oftennot parallel. The intensity and duration of the exer-cise appear to play a major role in this dichotomy of activity. After training, resting concentrations ofGH active in the rat bone growth assay are increased.3 The GH-producing cell system in the rat pituitarygland is heterogeneous. GH cellular heterogeneity mayalso be present in the human pituitary gland, but thesetypes of studies are difficult to do. Evidence supportsthe view that GH-containing secretion granules andGH-producing cells are heterogeneous. This hetero-geneity appears to have physiological relevance.Additional work is required to establish a linkbetween these components in exercising rats andhumans.4 Regulatory mechanisms responsible for releasing GHvariants from the pituitary may involve signals fromneural pathways originating in muscles activated byexercise. In this chapter we offer evidence to supportthe existence of a novel feedback loop from certainmuscle groups to the pituitary gland. This loop

appears to exist in humans as well as rats. It may bean important factor in controlling the productionand/or release of GH variants from the pituitarygland.

In conclusion, readers of this chapter know thatthe information explosion we are experiencing intoday’s biological sciences results not only frompast studies but from a rapidly expanding techno-logy and data base as well. This is obvious. In thischapter the authors have considered seminal worksdone some 50 years ago and tried to show that theyare relevant today and help in a fuller appreciationof the role that variant forms of the GH moleculemight play in the beneficial effects of human exer-cise. We have tried to show that a successful mar-riage between the experimental approaches used inendocrinology, endocrine biochemistry/cell biologyand exercise physiology may lead to new insightsinto the role that GH molecular heterogeneity playsin human exercise. Obviously a start has been made,but much more needs to be done.

Pretraining treatment groupsTibial line growth hormone

RT

Control

Pre-exercise

(a) (b)

Post-exercise

6000

5000

4000

3000

2000

1000

0Gro

wth

ho

rmo

ne

(ng

·mL–1

)

Post-training treatment groupsTibial growth hormone

Pre-exercise Post-exercise

6000

5000

4000

3000

2000

1000

0Gro

wth

ho

rmo

ne

(ng

·mL–1

)

RT

Control

Fig. 7.5 Bioassayable growth hormone in control versus exercise groups sampled pre and post an acute resistance exercisetest consisting of six sets of 10-RM squats separated by a 2-min rest period before (a) and after (b) 6 months of periodizedresistance training (RT) (unpublished data).

References

Baumann, G. (1991a) Growth hormoneheterogeneity: genes, isohormones,variants, and binding proteins. EndocrineReviews 12(4), 424–449.

Baumann, G. (1991b) Metabolism ofgrowth hormone (GH) and differentmolecular forms of GH in biologicalfluids. Hormone Research 36 (suppl. 1),5–10.

Baumann, G. (1999) Growth hormoneheterogeneity in human pituitary andplasma. Hormone Research 51 (suppl. 1),2–6.

Baumann, G., Shaw, M., Amburn, K. et al.(1994) Heterogeneity of circulatinggrowth hormone. Nucear Medicine andBiology 21(3), 369–379.

Bigbee, A.J., Gosselink, K.L., Roy, R.R.,

Grindeland, R.E. & Edgerton, V.R. (2000) Bioassayable growth hormonerelease in rats in response to a singlebout of treadmill exercise. Journal of Applied Physiology 89(6), 2174–2178.

Cunningham, B.C. & Wells, J.A. (1989)High-resolution epitope mapping of hGH-receptor interactions by

Page 108: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

92 chapter 7

alanine-scanning mutagenesis. Science244(4908), 1081–1085.

Cunningham, B.C., Ultsch, M., De Vos,A.M. et al. (1991) Dimerization of theextracellular domain of the humangrowth hormone receptor by a singlehormone molecule. Science 254(5033),821–825.

Dannies P.S. (1999) Protein hormonestorage in secretory granules:mechanisms for concentration andsorting. Endocrine Reviews 20(1), 3–21.

Ellis, S. & Grindeland, R.E. (1974)Dichotomy between bioassayable andimmunoassayable growth hormone. In: Advances in Human Growth HormoneResearch (Raiti, S., ed.). DHEWPublication No. (NIH) 74–612, USGovernment Printing Office,Washington, D.C.: 409–433.

Ellis, S., Nuenke, J.M. & Grindeland, R.E.(1968) Identity between the growthhormone degrading activity of thepituitary gland and plasmin.Endocrinology 83(5), 1029–1042.

Farrington, M. & Hymer, W.C. (1990)Growth hormone aggregates in the ratadenohypophysis. Endocrinology 126,1630–1638.

Gosselink, K.L., Grindeland, R.E., Roy,R.R. et al. (1998) Skeletal muscle afferentregulation of bioassayable growthhormone in the rat pituitary. Journal ofApplied Physiology 84(4), 1425–1430.

Gosselink, K.L., Grindeland, R.E., Roy,R.R. et al. (2000) Afferent input from ratslow skeletal muscle inhibitsbioassayable growth hormone release.Journal of Applied Physiology 88(1),142–148.

Greenspan, F.S., Li, C.H., Simpson, M.E. & Evans, H.M. (1949) Bioassay ofhypophyseal growth hormone: the tibiatest. Endocrinology 45, 455–463.

Hunter, W.M. & Greenwood, F.C. (1964) Aradio-immunoelectrophoretic assay forhuman growth hormone. BiochemistryJournal 91(1), 43–56.

Hymer, W.C. & McShan, W.H. (1963)Isolation of rat pituitary granules andthe study of their biochemical propertiesand hormonal activities. Journal of CellBiology 117(1), 67–86.

Hymer, W.C., Kirshnan, K., Kraemer, W.,Welsch, J. & Lanham, W. (2000)Mammalian pituitary growth hormone:applications of free flow electrophoresis.Electrophoresis 21(2), 311–317.

Hymer, W.C., Kraemer, W.J., Nindl, B.C. et al. (2001) Characteristics of circulatinggrowth hormone in women after acuteheavy resistance exercise. American

Journal of Physiology. Endocrinology andMetabolism 281(4), E878–E887.

Lewis, U.J., Pence, S.J., Singh, R.N. &VanderLaan, W.P. (1975) Enhancementof the growth promoting activity ofhuman growth hormone. Biochemical andBiophysical Research Communications67(2), 617–624.

Lewis, U.J., Sinha, Y.N. & Lewis, G.P.(2000) Structure and properties ofmembers of the hGH family: a review.Endocrine Journal 47 (suppl.), S1–S8.

McCall, G.E., Goulet, C., Grindeland, R.E. et al. (1997) Bed rest suppressesbioassayable growth hormone release in response to muscle activity. Journal ofApplied Physiology 83(6), 2086–2090.

McCall, G.E., Goulet, C., Roy, R.R. et al.(1999) Spaceflight suppresses exercise-induced release of bioassayable growthhormone. Journal of Applied Physiology87(3), 1207–1212.

McCall, G.E., Grindeland, R.E., Roy, R.R. & Edgerton, V.R. (2000) Muscle afferentactivity modulates bioassayable growthhormone in human plasma. Journal ofApplied Physiology 89(3), 1137–1141.

McCall, G.E., Gosselink, K.L., Bigbee, A.J.et al. (2001) Muscle afferent-pituitaryaxis: a novel pathway for modulatingthe secretion of a pituitary growth factor.Exercise and Sport Sciences Reviews 29(4),164–169.

Nguyen, N.Y., Grindeland, R.E. &Chrambach, A. (1981) Isolation ofhuman growth hormone isohormones D and E in milligram amounts (II), usingisoelectric focusing on polyacrylamidegel. Preparative Biochemistry 11(2),173–189.

Nindl, B.C., Kraemer, W.J. & Hymer, W.C. (2000) Immunofunctional vs.immunoreactive growth hormoneresponses after resistance exercise inmen and women. Growth Hormone andIGF Research 10(2), 99–103.

Nindl, B.C., Hymer, W.C., Deaver, D.R. &Kraemer, W.J. (2001) Growth hormonepulsatility profile characteristicsfollowing acute heavy resistanceexercise. Journal of Applied Physiology91(1), 163–172.

Nindl, B.C., Kraemer, W.J., Marx, J.O.,Tuckow, A.P. & Hymer, W.C. (2003)Growth hormone molecularheterogeneity and exercise. Exercise andSport Sciences Reviews 31(4), 161–166.

Paden, C., Moffett, C. & Benowitz, L. (1994) Innervation of the rat anterior and neurointermediate pituitaryvisualized by immunocytochemistry for the growth-associated protein

GAP-43. Endocrinology 134(1), 503–506.

Papkoff, H. & Li, C.H. (1962) Hypophysealgrowth hormone. In: Methods in HormoneResearch II (Dorfman, R. ed.). AcademicPress, New York: 671–704.

Roswall, E.C., Mukku, V.R., Chen, A.B. et al. (1996) Novel assays based onhuman growth hormone receptor asalternatives to the rat weight gainbioassay for recombinant human growthhormone. Biologicals 24(1), 25–39.

Roth, J., Glick, S.M., Yalow, R.S. & Berson,S.A. (1963) Secretion of human growthhormone: physiologic and experimentalmodification. Metabolism 12, 577–579.

Rowlinson, S.W., Waters, M.J., Lewis, U.J.& Barnard, R. (1996) Human growthhormone fragments 1–43 and 44–191: invitro somatogenic activity and receptorbinding characteristics in human andnonprimate systems. Endocrinology137(1), 90–95.

Rubin, M.R., Kraemer, W.J., Kraemer, R.R.et al. (2003) Responses of growthhormone aggregates to differentintermittent exercise intensities.European Journal of Applied Physiology89(2), 166–170.

Singh, R.N., Seavey, B.K., Rice, V.P.,Lindsey, T.T. & Lewis, U.J. (1974)Modified forms of human growthhormone with increased biologicalactivities. Endocrinology 94(3), 883–891.

Stolar, M.W., Amburn, K. & Baumann, G.(1984) Plasma ‘big’ and ‘big–big’growth hormone (GH) in man: anoligomeric series composed ofstructurally diverse GH monomers.Journal of Clinical Endocrinology andMetabolism 59(2), 212–218.

Strasburger, C.J. (1994) Implications ofinvestigating the structure–functionrelationship of human growth hormonein clinical diagnosis and therapy.Hormone Research 41 (suppl. 2), 113–119.

Strasburger, C.J., Wu, Z., Pflaum, C.D. & Dressendorfer, R.A. (1996)Immunofunctional assay of humangrowth hormone (hGH) in serum: apossible consensus for quantitative hGH measurement. Journal of ClinicalEndocrinology and Metabolism 81(7),2613–2620.

Wada, M., Uchida, H., Ikeda, M. et al.(1998) The 20-kilodalton (kDa) humangrowth hormone (hGH) differs from the22-kDa hGH in the complex formationwith cell surface hGH receptor andhGH-binding protein circulating inhuman plasma. Molecular Endocrinology(Baltimore, Md) 12(1), 146–156.

Page 109: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone variants and human exercise 93

Wallace, J.D., Cuneo, R.C., Bidlingmaier,M. et al. (2001) The response ofmolecular isoforms of growth hormoneto acute exercise in trained adult males.Journal of Clinical Endocrinology andMetabolism 86(1), 200–206.

Experimental Biology and Medicine 159,409–413.

Wood, P. (2001) Growth hormone: itsmeasurement and the need for assayharmonization. Annals of ClinicalBiochemistry 38(5), 471–482.

Weiss, S., Bergland, R., Page, R., Turpen, C.& Hymer, W.C. (1978) Pituitary celltransplants to the cerebral ventriclespromote growth of hypophysectomizedrats. Proceedings of the Society for

Page 110: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

94

Growth hormone binding proteins (GHBPs) are soluble, circulating proteins that form complexeswith growth hormone (GH). They are integral partsof the somatotropic axis and have activities as mod-ulators of GH action and GH transport in blood. An overview of the growth hormone–insulin-likegrowth factor (GH–IGF) axis is shown in Fig. 8.1.

History

The presence of GHBPs in blood was first postu-lated in the 1960s (Irie & Barrett 1962; Touber &Maingay 1963; Collipp et al. 1964; Hadden & Prout1964), but these observations were not generallyaccepted as physiological phenomena at that time(Berson & Yalow 1966a, 1966b). In 1977, Peeters &Friesen (1977) described a GH binding factor inpregnant mouse serum. However, this observationwas also largely ignored. It was not until Baumannand Herington independently described, character-ized and partially purified GHBPs from human andrabbit serum (Ymer & Herington 1985; Baumann et al.1986; Herington et al. 1986b) that the GHBPs weregenerally accepted as authentic. Two GHBPs, onewith high affinity and the other with low affinity forGH were described at that time (Baumann et al.1986). While the high affinity GHBP was easy towork with and soon became recognized as the GHreceptor (GHR) ectodomain (Leung, D.W. et al. 1987;Baumann et al. 1988), it took some years to character-ize the low affinity GHBP (Baumann et al. 1990; Taret al. 1990), ultimately leading to its recognition astransformed α2-macroglobulin (Kratzsch et al. 1995b).In general, the term ‘GHBP’ is used for the high

affinity GHBPaa convention that will also be fol-lowed in this chapter unless stated otherwise.

Nature and chemical properties

The high affinity GHBP represents the extracellulardomain of the GHR (Leung, D.W. et al. 1987; Spenceret al. 1988). It is a single chain glycoprotein with amolecular weight that varies widely depending on the species from 28 kDa (chicken) to 65 kDa(humans), with most of the variation due to differ-ences in glycosylation. The polypeptide backboneaccounts for approximately 28–30 kDa, with minordifferences among species. The GHBP is evolution-arily conserved from teleost to humans; it has beenfound in the blood of all vertebrates examined. Insome species it is derived directly from the GHR byproteolysis, in others (rodents), it is synthesized as a separate gene product (see below). The precisestructure of the GHBP is known in only a few spe-cies, with the carboxy terminus unknown in many.Two subdomains, each composed of beta-pleatedsheets, have been identified: an amino-terminal subdomain 1 containing the GH binding site; and acarboxy-terminal subdomain 2 that is involved indimerization of the GHR. An approximately 10amino acid linear stem region extends between sub-domain 2 and the transmembrane helix of the GHR(Baumann & Frank 2002). The precise cleavage sitein the GHR giving rise to the GHBP has recentlybeen mapped in the rabbit: cleavage occurs in theextracellular stem region between subdomain 2 andthe transmembrane domain, with the GHBP car-boxy terminus at residue no. 238, i.e. eight residues

Chapter 8

Growth Hormone Binding Proteins

GERHARD BAUMANN

Page 111: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 95

outside the plasma membrane (Wang et al. 2002).Based on the sequence similarity of the rabbit andhuman GHRs in the extracellular stem region, itappears likely that the human GHBP has the samelength, though this still needs direct confirmation.In rodents, the GHBP is the product of an altern-atively spliced GHR mRNA and is synthesized

de novo. It contains a carboxyterminal ‘tail’ of 27(mouse) or 17 (rat) amino acids in lieu of the trans-membrane domain in the GHR (Baumbach et al.1989; Smith et al. 1989). The mouse and rat GHBPcontain 273 and 255 amino acids, respectively. Theextent of GHBP glycosylation differs among spe-cies, but there is only limited information for rodentGHBP about the nature of the sugar moieties.Mouse serum GHBP is glycosylated at three aspara-gine residues, wheras tissue-associated GHBP (seebelow) contains less carbohydrate and is glyco-sylated at two asparagine residues (Cerio et al.2002). Rat serum GHBP contains sialic acid, whereastissue-associated GHBP is rich in mannose (Frick et al. 1998). The details of the carbohydrate sidechain structure are not known. In humans, two highaffinity GHBPs exist (one containing and the otherlacking the sequence encoded by exon 3 of the GHRgene) (Kratzsch et al. 1997b). This is the consequenceof GHR polymorphism with respect to exon 3(Pantel et al. 2000; Seidel et al. 2003). The presence orabsence of the exon 3-encoded sequence in the GHRor the GHBP has no significant functional con-sequence with respect to GH binding. However,small differences in the correlations between serumGHBP level and anthropometric/metabolic para-meters have been reported for the two GHBP iso-forms (Seidel et al. 2003).

The high affinity GHBP binds GH with dissoci-ation constants ranging from 10–8 to 10–9 mol (Ymer & Herington 1985; Baumann et al. 1986; Herington et al. 1986b; Smith et al. 1988; Massa et al. 1990). It exhibits somewhat lower affinity (Kd 10–6 to 10–7)for the 20 000 kDa (20 K) variant of human GH(Baumann et al. 1986). Like the GHR, the GHBP hasthe capacity to form ternary (2 GHBP : 1 GH) com-plexes with GH, but due its low concentration inbiological fluids, the 1 : 1 GHBP–GH complex pre-dominates under physiological conditions (Baumannet al. 1994). The association rate for the humanGHBP is very rapid (~ 2 × 107·mol−1·min−1 at 37°C,80% maximum binding reached in 5 min), the dis-sociation rate is 3.7 × 10−2·min−1 at 37°C (disso-ciation half-time ~ 20 min) (Baumann et al. 1986;Veldhuis et al. 1993, Baumann 1995).

The low affinity GHBP is a heterogeneous plasmacomponent that binds GH with a Kd in the micromolar

Hypothalamus

GHRH (+)

GHBP

(−)(−)

Somatostatin (−)

Pituitary

GH

Other tissuesLiver

Local IGF-ICirculating IGF-I

Growth/Anabolism

(−)

Fig. 8.1 The hypothalamo–pituitary–somatotropic axis,also known as growth hormone–insulin-like growthfactor (GH–IGF) axis. Pituitary GH secretion is underpositive (stimulatory) control from the hypothalamus viagrowth hormone releasing hormone (GHRH) and undernegative (inhibitory) control via somatostatin. Aftersecretion, GH binds to receptors in liver and virtually allother tissues, and to growth hormone binding proteins(GHBPs) in the circulation. There is interchange betweenfree GH, GHBP-bound GH and receptor-bound GH.Peripheral tissues produce IGF-I in response to GH. Theliver is responsible for 60–70% of circulating IGF-I; othertissues are responsible for the rest. In tissues other thanthe liver, local (paracrine/autocrine) IGF-I action is veryimportant. Direct (non-IGF-dependent) GH actions are indicated by the dashed arrow. IGF-I feeds backnegatively on the hypothalamus and pituitary gland to inhibit GH secretion. GH itself also inhibits its ownproduction via negative feedback at hypothalamic level(short-loop feedback).

Page 112: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

96 chapter 8

range (Baumann et al. 1986, 1990; Massa et al. 1990;Tar et al. 1990; Leung, K.C. et al. 2000). It has highbinding capacity and in humans has been shown torepresent a modified form of α2-macroglobulin(‘transformed α2-macroglobulin’) (Kratzsch et al.1995b). Relatively little is known about the mole-cular nature of low affinity GHBPs in animals.

Generation and tissues source(s)

As indicated above, the high affinity GHBP is gener-ated by different mechanisms depending on spe-cies. In humans, rabbits and several other species,the GHBP is produced by proteolytic cleavage of the juxtamembranous stem region of the GHR, aprocess named ‘shedding’ (Fig. 8.2). The enzymeinvolved in this process has been recently identified:it is a zinc metalloproteinase of the ADAM familynamed TACE (tumor necrosis factor-α convertingenzyme), also known as ADAM-17 (Black et al. 1997;Zhang et al. 2000). Mature, catalytically active TACEis a plasma membrane-resident enzyme that asso-ciates with the GHR, followed by cleavage andshedding of GHBP and a truncated GHR ‘remnant’,which has its own intracellular fate. TACE isresponsible for cleavage of a number of transmem-brane proteins in their extracellular domain, withshedding of soluble ectodomains akin to the GHBP.

It is possible that other enzymes in the same classalso contribute to GHBP shedding, but this remainsto be investigated. The regulation of TACE activityand the shedding process are still poorly under-stood. The conformational change induced in theGHR by binding of GH (dimerization or change inpredimerized GHR) renders it less prone to proteo-lysis than the monomeric, unliganded GHR (Zhanget al. 2001). GHBP shedding is thought to occur prin-cipally if not exclusively at the cell surface, based onthe localization of active TACE and the fact that aGHR variant with a long plasma membrane residencetime (devoid of the cytoplasmic/internalizationdomain) is a particularly good source of GHBP(Dastot et al. 1996).

Rodents generate GHBP by an entirely differentmechanism. Both mouse and rat ghr genes contain aspecial exon (exon 8A) encoding the hydrophilicGHBP tail (see above) interposed between exons 7and 8 (Edens et al. 1994; Zhou et al. 1994, 1996). Exon8 encodes the transmembrane helix. DifferentialmRNA splicing of exon 7 to either exon 8A or exon 8yields the GHBP or the GHR, respectively (Fig. 8.3).Both transcripts are expressed in the same tissues,but it is unknown what regulates their relativeexpression. It should be noted that the mouse GHRis not completely resistant to TACE proteolysis, atleast when induced by phorbol ester. However, the

Zn GHBP

Circulation

Plasma membrane

Further processingInternalizationDegradation

GHRremnant

GHR

TACEPKCMAPK etc.

Fig. 8.2 The generation of growthhormone binding protein (GHBP) via proteolytic cleavage of the growth hormone receptor (GHR). Amembrane-bound zinc-dependentmetalloproteinase (tumor necrosisfactor-α converting enzyme [TACE]) cleaves the GHR in itsjuxtamembranous stem region 8–9amino acid residues outside theplasma membrane. The GHRectodomain is shed as the GHBP,which reaches the circulation. TheGHR remnant protein, consisting ofthe transmembrane and intracellulardomains, undergoes additionalprocessing and may have its ownbiological role. TACE is activated byMAP kinase (MAPK) and proteinkinase C (PKC) dependent pathways.

Page 113: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 97

cleavability of the mouse GHR is about two ordersof magnitude lower than that of the rabbit GHR (G.Baumann, unpublished). In vivo, it appears thatmost if not all of the circulating rat GHBP is derivedfrom the alternative mRNA splicing mechanism(Sadeghi et al. 1990). The two different mechanismsof GHBP generation are illustrated in Fig. 8.4.

Rhesus monkeys use both the proteolytic and an alternative mRNA splice mechanism for GHBPgeneration (Martini et al. 1997). In that case, thealternative mRNA encoding the GHBP derives froma readthrough into intron 7, which results in a 7amino acid ‘tail’ replacing the transmembranedomain due to an intronic stop codon. It is notknown which mechanism predominates in generat-ing the GHBP in the monkey.

The tissue sources of the GHBP are well-definedin the rodent, where the GHBP can be specificallyrecognized and differentiated from the GHR at both the mRNA and protein level by its unique carboxyterminal tail. GHBP is expressed ubiquit-ously, and generally is coexpressed with the GHR(Carlsson, B. et al. 1990; Lobie et al. 1992). However,their expression is not necessarily regulated in aparallel fashion (Walker et al. 1992). Of interest, a

substantial portion of rodent GHBP remains associ-ated with the plasma membrane (and intracellularmembranes) by an as yet unknown link (Frick et al.1994, 1998); it has been suggested that an Arg-Gly-Asp sequence in the GHBP may interact withmembrane integrins to provide a tether (Cerio et al.2002). GHBP in the circulation differs from the tissue-associated form in its glycosylation moiety.Membrane-associated GHBP forms have not beendescribed in non-rodent species. The tissue sourceof the GHBP in species employing the proteolyticshedding mechanism is less clear as it is moredifficult to differentiate the GHBP from the GHR.Since both the GHR and TACE are expressed ubi-quitously, all tissues can theoretically contribute to GHBP generation. However, the quantitativeaspects of GHBP generation by individual tissuesare not clearly established. Based on the relativeabundance of GHRs in the liver, that organ is gen-erally thought to be a major source. However, itshould be noted that this concept has not beendirectly validated. Studies of venous gradients invisceral organ effluents have not identified a majororgan source (Segel et al. submitted for publication).It appears likely that multiple tissues contribute

Human GHR gene

GHR mRNA

2

2

3

3

4

4

5

5

6

6

7

7

8

8

8a

9

9

10

10

2 3 4 5 6 7 8a

Mouse ghr gene

GHBP mRNA

Fig. 8.3 The human and mouse GHR genes and their products are shown. In humans and most other species, the onlygene product is the growth hormone receptor (GHR). In rodents, an additional embedded exon (exon 8a) encodes thehydrophilic tail of the growth hormone binding protein (GHBP). Splicing exon 8a to exons 2–7 yields the GHBP mRNA(bottom). Splicing exon 8–10 to exons 2–7 yields the GHR mRNA (top). Exon 8a is not spliced to downstream exonsbecause it contains a cleavage/polyadenylation site and lacks a canonical splice donor site at its 3’ end. Exon 1 of the GHRgene is not shown because it is not part of the coding region (it encodes the 5’ untranslated region[s]). Light tint denotesthe extracellular, darker tint the intracellular domain. denotes the transmembrane domain, the hydrophilic GHBP tail.

Page 114: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

98 chapter 8

to the circulating GHBP pool, though their relativecontributions remain to be determined.

Growth hormone binding proteins inbiological fluids

The high affinity GHBP is found in blood and mostother biological fluids, such as urine, lymph, milk,semen, follicular fluid and amniotic fluid (Hattori et al. 1990; Postel-Vinay et al. 1991a; Amit et al. 1993;Maheshwari et al. 1995; Harada et al. 1997).Cerebrospinal fluid contains no detectable GHBP(Nixon & Jordan 1986). Unlike in rabbit milk, theGHBP found in human milk appears to related to theprolactin receptor rather than the GHR (Mercado & Baumann 1994). The concentration of GHBP inblood varies over a 10-fold range; it is generally pre-sent at nanomolar to subnanomolar concentrations.

This concentration, together with its affinity, allowsthe GHBP to act as a buffer and dynamic modulatorfor circulating free GH. Under physiological andbasal conditions, approximately 45% of circulatingGH in human blood is bound to the high affinityGHBP (Baumann et al. 1988, 1990). This proportionchanges dynamically after a GH secretory spike(Veldhuis et al. 1993).

GHBP is also present within the cell (Herington et al. 1986a; Lobie et al. 1991; Frick et al. 1994), but thesource, destination and function of this intracellularGHBP is not clear.

The low affinity GHBP has only been described inblood, where it circulates at micromolar concentra-tions (Baumann et al. 1990; Leung, K.C. et al. 2000). Inhumans, approximately 8% of circulating GH isbound to this GHBP; in rats it can be calculated thatabout 20% of GH is bound to the low affinity GHBP

Proteolyticcleavage

COOH

(a)

Receptor mRNA

NH2

Humans, rabbit,other species

COOH

(b)

Receptor mRNA

NH2

BP mRNA

Mouse, rat

Secretion

Binding proteinReceptor

Fig. 8.4 The two mechanisms of growth hormone binding protein (GHBP) generation are shown. (a) The proteolyticshedding mechanism. (b) The splice variant/direct synthesis and secretion mechanism. (Adapted from Baumann, G.(1990) Growth hormone binding proteins. Trends in Endocrinology and Metabolism 1, 342–347. Copyright 1990, withpermission from Elsevier.)

Page 115: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 99

(Barsano & Baumann 1989; Baumann et al. 1989a;Leung, K.C. et al. 2000).

Functional aspects

The principal known function of the GHBPs is toform complexes with GH. Quantitatively, this ismore important for the high affinity GHBP than thelow affinity GHBP. An indirect ‘function’ of GHBPgeneration is the inactivation of GHRs by cleavingand shedding its ectodomain, a process that can beviewed as receptor ‘decapitation’. GH binding hasmultiple consequences. At the local (cellular/tissue)level, GHBP competes with the GHR for GH ligand,the consequence of which is decreased GH action(Fig. 8.5). This effect can be clearly shown in vitro,where GHBP inhibits GH binding to GHRs and GHaction in a dose-dependent manner (Lim et al. 1990;Mannor et al. 1991). An additional likely reason for

decreased GH action is the formation of unproduct-ive, non-signaling GHR/GHBP dimers (Fig. 8.5).Dimerization and proper GHR dimer conformationis necessary for signal transduction by the GHR. AGHR/GHBP dimer cannot perform that function.Formation of such heterodimers by GH bindingwould inhibit GH action in a GHBP concentration-dependent fashion. Indeed, this effect has beendemonstrated for naturally occurring and mutantforms of the GHR that lack the intracellular domain(Ayling et al. 1997; Ross et al. 1997; Iida et al. 1999). Ithas not directly been proven that the same phe-nomenon occurs with soluble GHBP, though thiswould be predicted. The short GHR, in contrast tothe GHBP, contains a transmembrane domain and is membrane-anchored. If the GHR exists in themembrane in a predimerized form even in theabsence of GH binding (Ross et al. 2001), the mem-brane-resident short GHR form is not necessarily

GHBP

GHR GHR GHR

GH

GHR GHBP

GH

GH

Jak2 Jak2

Intracellularsignaling

(a) (b) (c)

Plasma membrane

Fig. 8.5 Inhibitory actions of the growth hormone binding protein (GHBP) on GH bioactivity. (a) GHBP competes withthe GHR for GH. (b) Normal GH signaling through the growth hormone receptor (GHR). Binding of GH to the GHRinduces GHR dimerization or activation of a preformed GHR dimer. The conformationally appropriate GHR-dimer bindsJak2 and initiates intracellular signaling. (c) A GHR–GHBP heterodimer is unable to initiate signaling, with the GHBPacting as a dominant negative inhibitor.

Page 116: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

100 chapter 8

representative for the extracellular, soluble GHBP.Therefore, the concept of GHR/GHBP heterodimersremains to be formally validated.

In contrast to its inhibitory action in vitro, theGHBP tends to enhance GH action in vivo. GHBPprolongs the plasma half-life of GH through forma-tion of a complex that is too large for efficient glomer-ular filtration and renal eliminationathe principalroute of GH clearance (Baumann et al. 1987a, 1989b).The complex also diminishes GH clearance throughGHR-mediated mechanisms, such as cellular inter-nalization, and delays chemical degradation. In rats,the metabolic clearance of complexed GH is 10-foldlower than that of free GH (Baumann et al. 1989b). In humans, the GH–GHBP complex has been estim-ated to have a plasma half-life of 25–29 min, versus4–9 min for free GH (Veldhuis et al. 1993). TheGH–GHBP complex in blood serves as a circulatingGH reservoir, which dynamically dampens GHoscillations resulting from secretory pulses. GHBP,when given in large doses, has been shown toenhance GH bioactivity in vivo, despite its inhibitoryactions in vitro (Clark et al. 1996). The net effect of the high affinity GHBP on GH action in the in-tact organism is thus complex, concentration- andcompartment-dependent, and difficult to predict.

Little is known about the effect of the low affinityGHBP on GH action. Based its low affinity, it islikely to form a loose complex with GH that easily dissociates. Therefore, it probably has only a limitedimpact on GH dynamics and GH action.

Regulation of growth hormone binding protein production

In species that generate GHBP by the proteolyticmechanism, GHBP production depends both onGHR expression and on the regulation of TACEactivity. Both the GHR and TACE are expressedubiquitously. GHR expression is regulated bydevelopmental stage, gender, species, metabolicstate, and differentially by tissue. Little is currentlyknown about the regulation of TACE activity. Inrodents, GHBP production is linked to expression of the GHBP mRNA variant. This is also regulatedin a complex, tissue and metabolic state-dependentmanner, and no systematic studies exist that com-

prehensively address this issue. Because of theselimitations, the following discussion is primarilyfocused on the regulation of GHBP levels in serum.

In humans, the principal factors affecting serumGHBP as part of normal physiology are develop-ment, gender, aging and nutrition. For reasons thatare not known, serum GHBP concentrations in normal subjects vary over a 10-fold range (~ 0.3–3.0 nmol) (Rajkovic et al. 1994; Maheshwari et al.1996); it is not clear whether this normal variablityhas biological significance. There is no significantdiurnal variation in serum GHBP (Snow et al. 1990;Carmignac et al. 1992; Carlsson, L.M. et al. 1993), buta minor seasonal variation with nadir in August has been reported in children (Gelander et al. 1998).GHBP levels are very low in the fetus, rise rapidlyduring early childhood, stay constant through ado-lescent and adult life, and decline in old age after the age of 60 years (Daughaday et al. 1987; Holl et al.1991; Martha et al. 1993; Maheshwari et al. 1996). Asimilar ontogenetic pattern is present in the rat(Mulumba et al. 1991). GHBP levels are higher infemales than males, both in humans and moremarkedly in rodents (Massa et al. 1990; Hattori et al.1991; Rajkovic et al. 1994). This is probably in largepart an estrogen effect. Maternal GHBP levels dur-ing pregnancy show a very marked species differ-ence. In humans, there is only a small increase inGHBP levels in early pregnancy (Blumenfeld et al.1992), whereas in mice, GHBP levels in serum (andmembrane-associated GHBP in the liver) show avery large increase (Cramer et al. 1992; Camarillo et al. 1998). It is the latter phenomenon that led to the first description of the GHBP (Peeters & Friesen1977). Rats also increase their GHBP during preg-nancy, though to a lesser degree than mice (Frick et al. 1998). An important factor regulating GHBPlevels is nutrition. Malnutrition decreases and over-nutrition increases serum GHBP; and a highly sig-nificant correlation exists between body mass indexand GHBP levels, and especially between visceralfat and GHBP levels (Hochberg et al. 1992; Martha et al. 1992; Roelen et al. 1997b). These changes paral-lel those seen for IGF-I levels and probably reflectthe effect of insulin on GHR expression and conse-quently GHBP levels (Baxter & Turtle 1978; Mercadoet al. 1992; Kratzsch et al. 1996).

Page 117: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 101

GH up-regulates GHBP in rodents (Sanchez-Jimenez et al. 1990; Carmignac et al. 1992), but dataon this subject in humans are variable and incon-sistent (see Baumann 2001 for review). It may beconcluded from this that GH does not have a majoreffect on GHBP levels in humans. Of interest,acromegaly, a disease of chronic GH excess, is associated with low to low normal GHBP in moststudies (Amit et al. 1992; Roelen et al. 1992; Mercadoet al. 1993; Kratzsch et al. 1995a; Fisker et al. 1996).This is not necessarily a direct function of GH, butmay be a result of other adjustments occurring inacromegaly. Thyroid hormone up-regulates GHBPlevels (Amit et al. 1991; Romero et al. 1996).Estrogens, especially when given by the oral route,increase GHBP levels in humans and rodents, butlower serum GHBP in the rabbit (Weissberger et al.1991; Carmignac et al. 1993; Yu et al. 1996). Andro-gens lower serum GHBP levels (Postel-Vinay et al.1991b; Keenan et al. 1996; Yu et al. 1996). Glucocor-ticoids lower GHBP in humans and rodents, butincrease GHBP in rabbit blood (Heinrichs et al. 1994;Miell et al. 1994; Gabrielsson et al. 1995). Insulin up-regulates GHBP levels (Mercado et al. 1992; Massa et al. 1993; Kratzsch et al. 1996), whereas IGF-I, in a single report, was shown to lower GHBP levels(Silbergeld et al. 1994).

Exercise and physical training have an effect onplasma GHBP levels. Acute exercise, such as cycleergometry, induces a short-lived and mild increasein GHBP (Wallace et al. 1999). Chronic endurance orfitness training has been shown to lower serumGHBP by 10–40% in most studies (Roemmich &Sinning 1997; Eliakim et al. 1998b, 2001; Scheett et al.2002), but at least one study showed a small rise inGHBP (Roelen et al. 1997a). GHBP is inversely relatedto peak oxygen uptake and fitness (Eliakim et al.1998a). This is in part linked to the above-mentionedrelatienship between adiposity and GHBP. Thephysiological significance of these exercise/trainingrelated GHBP changes is not fully understood.

Growth hormone binding protein and disease

Several pathological conditions are associated withabnormal serum GHBP levels. In most cases, the

change in GHBP parallels altered GH sensitivity;the GHBP level is therefore thought to mirror GHRabundance in tissues. Foremost among the disor-ders with abnormal GHBP is the genetic GH insens-itivity syndrome due to inactivating mutations inthe GHR gene (Laron syndrome), which results insevere growth retardation and dwarfism (Rosenfeldet al. 1994). Absence or malfunction of the GHBP isgenerally a direct result of the mutant GHR, whichis either not expressed (e.g. gene deletions, non-sense mutations), prematurely degraded or notproperly directed to the plasma membrane (e.g.some missense mutations), or unable to bind GH(certain missense mutations) (see Baumann 2002 foran updated listing of known GHR mutations).Absence of GHBP activity in the serum of patientswith Laron syndrome was the first strong indicationthat the GHBP is a GHR fragment (Baumann et al.1987b; Daughaday & Trivedi 1987). About 80% ofcases with Laron syndrome have low or undetect-able GHBP in their blood (Woods et al. 1997). Theothers have either normal serum GHBP or in rarecases even elevated GHBP. The underlying GHRmutations in GHBP-positive cases include thosethat prevent proper receptor dimerization or lackthe intracellular signaling domain (Duquesnoy et al.1994; Ayling et al. 1997; Kaji et al. 1997; Iida et al.1998; Gastier et al. 2000). A mutant receptor lackingthe transmembrane helix results in massively elev-ated serum GHBP activity, which represents themutant, soluble GHR rather than the normal GHBP(Woods et al. 1996; Silbergeld et al. 1997).

Several disorders associated with acquired GHinsensitivity are also characterized by abnormallylow GHBP levels. Catabolic conditions, such as malnutrition, insulinopenic uncontrolled diabetes,post-traumatic states and critical illness are examplesof acquired GH-resistance, characterized by lowIGF-I levels despite normal or elevated GH secre-tion. Growth failure may occur in severe cases(Mauriac syndrome, a condition of poorly con-trolled diabetes, hepatomegaly and growth retarda-tion) (Mandell & Berenberg 1974; Mauras et al.1991). The fact that serum GHBP is decreased inGH-resistant states lends credence to the conceptthat serum GHBP concentration reflects tissue GHRabundance. In animal models, catabolic states are

Page 118: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

102 chapter 8

associated with decreased hepatic GHR levels andGH resistance (Baxter & Turtle 1978; Postel-Vinay et al. 1982; Massa et al. 1993). Upon resolution of theunderlying disease process, GH sensitivity, GHRexpression and GHBP levels return to normal. Thechanges in GHR expression and consequent GHBPrelease are thought to be largely mediated by insulin(Mercado et al. 1992; Hanaire-Broutin et al. 1996).

The counterpart of GH resistance, GH hyper-sensitivity, is associated with elevated GHBP levels.The only well-recognized condition in this categoryis overnutrition/obesity, which is characterized bynormal or elevated IGF-I levels despite suppressedGH secretion. It has long been known that over-weight children grow faster than lean children(Forbes 1977). Obesity is associated with elevatedserum GHBP levels, probably reflecting increasedtissue GHR expression (Hochberg et al. 1992;Kratzsch et al. 1997a; Roelen et al. 1997b). Thus, the biochemical parameters as well as the functionalaspects of the GH–IGF axis in obesity are the exactopposite from those in malutrition.

With regard to the low affinity GHBP, little isknown about changes in its serum level in eithernormal physiology or in disease.

Assays for the growth hormone bindingprotein (Figs 8.6 and 8.7)

The classical assay for both the high and the lowaffinity GHBPs measures their functional propertyof radiolabeled GH binding, with separation ofbound from free GH by size exclusion chromato-graphy (Baumann et al. 1986; Herington et al. 1986b).This assay is quantitative under most circumstancesbecause under physiological conditions the GHBPsin serum are largely unoccupied. Correction foroccupation of the high affinity GHBP by endogen-ous GH must be made above a GH concentration of 10 ng·mL–1 (Baumann et al. 1989a). Variations ofthis fundamental GH binding assay employ othermethods for separation of bound from free GH,such as charcoal or immunoprecipitation with anti-GHR antibodies (Barnard et al. 1989; Amit et al. 1990;

GHIncubate

8

6

4

125 I

(cp

m x

10-3

/fra

ctio

n)

2

050 100

Fraction number

Size exclusionchromatography

Separate free from bound GH

Immunoprecipitationof bound GH

Charcoal adsorptionof free GH

IVI

II

Vo

III

150

125I-GH

GHBP

Vt Fig. 8.6 Growth hormone bindingprotein (GHBP) assays based on theprinciple of GH binding. To serumcontaining GHBP and endogenousGH radioiodinated GH is added, and GHBP–GH complex formation is allowed. Bound GH is thenseparated from free GH by either size exclusion chromatography (top),immunoprecipitation with anti-GHantibodies (center), or adsorption offree GH with activated charcoal(bottom). The assay is quantitativebecause under most conditions,GHBP is essentially unoccupied by endogenous ligand. At highendogenous GH levels (> 10ng·mL–1), correction for GHBPsaturation by unlabeled GH isnecessary. Different variations of thisbinding assay exist, using othermodes of separating free from boundGH.

Page 119: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 103

Coat plate with anti-GHR/GHBP antibody

Add serum sample and GH. Incubate

Anti-GH antibody

Enzyme

GHBP

GH

Add enzyme-coupled anti-GH antibody.Develop with substrate → Colorimetry

Fig. 8.7 Ligand-mediated immunofunctional assay (LIFA) for growth hormone binding protein (GHBP). This is a solid-phase, sandwich-type, two-site assay. A monoclonal antibody directed against the extracellular domain of the growthhormone receptor (GHR) (the GHBP), yet not interfering with its GH-binding site, is adsorbed to a microtiter plate. After washing, serum and an excess of exogenous GH are added, and formation of a ternary complex consisting of theantibody, GHBP and GH is allowed to form. After washing, an anti-GH-antibody coupled to an enzyme (e.g. horseradishperoxidase) is added. The antibody recognizes the solid-phase complex. Signal amplification is then achieved by theenzymatic reaction, using a color-yielding substrate.

Ho et al. 1993). Specific assays that differentiate theGHBP from the GHR have been devised by usingantibodies directed against the unique hydrophilictail of the rodent GHBP (Barnard et al. 1994); thisstrategy cannot be used for GHBP in species that usethe proteolytic shedding mechanism for GHBP gen-eration (e.g. humans, rabbits). A two-site sandwich-type assay with enzyme linked immunosorbentassay (ELISA) design (‘ligand-mediated immuno-functional assays [LIFAs]’) has been developed forthe human high affinity GHBP (Carlsson, L.M. et al.1991). Its results correlate well with the conven-tional GH-binding assay, but for unknown reasonsthe absolute GHBP values are lower than thoseobtained in other assays (Mercado et al. 1993). Oneassay with classical radioimmunoassay design(radiolabeled GHBP tracer and anti-GHBP anti-serum) has been reported for human GHBP(Kratzsch et al. 1995a). This type of assay is independ-ent of GH binding and able to measure dysfunc-tional GHBP with abnormal GH binding properties(such as mutant GHBP in certain cases of Laron syn-drome). A radioimmunoassay specific for the exon3-containing human GHBP has also been developed

(Kratzsch et al. 1997b). Other assays with variationson these methodological themes have been reportedfor several species, including human, rat and mouseGHBP. Unfortunately, relatively little information isavailable about the correlation among these assays.Commercial assays are available, but also sufferfrom a relative lack of crossvalidation with estab-lished assays. GHBP measurement is still primarilya research tool; its main practical application in clin-ical medicine is in the diagnosis of GH insensitivity(Laron) syndrome.

No standardized assays for the low affinity GHBPare available. This plasma component has beenmeasured by GH binding assay followed by sizeexclusion chromatography (Baumann et al. 1989a;Tar et al. 1990) or immunoprecipitation with anti-α2-macroglobulin antibody (Kratzsch et al. 1995b).

Effect of the high affinity growth hormonebinding protein on growth hormonemeasurement in serum

The high affinity GHBP can interfere with GH im-munoassays in serum because of competition with

Page 120: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

104 chapter 8

antibodies for GH. In general, antibodies, especi-ally when polyclonal, have higher affinity for GHthan the GHBP. Nevertheless, depending on theassay design, interference can be significant. Assaysparticularly vulnerable are those employing relat-ively low affinity monoclonal anti-GH antibodies,quick turnaround assays that are run under briefincubation, non-equilibrium conditions and insens-itive assays employing a high serum volume. Thenon-equilibrium assay design is problematic becausetime is required to transfer GHBP-bound GH fromthe GHBP to the antibody. Reports of interference in GH assays by the GHBP range from negligible to significant (Jan et al. 1991; Chapman et al. 1994;Jansson et al. 1997; Fisker et al. 1998). It is importantto carefully examine each GH immunoassay forGHBP interference as not all factors responsible forinterference are known, and assay methodologyshould be optimized to minimize GHBP effects.

Conclusions

Two GHBPs exist in blood and other biologicalfluids. The high affinity GHBP is the ectodomain of the GHR; it is either shed from cells by the actionof TACE, a member (ADAM-17) of the ADAM family of metalloproteinases, or directly secreted (in

rodents) as a soluble GHR mRNA splice variant.Cleavage by TACE occurs in the juxtamembranous,extracellular stem region of the GHR. The GHBP has complex effects on GH blood transport, GHclearance and GH action, with both enhancing andinhibitory modulation of GH bioactivity. The physio-logical significance of the GHBP for the somato-tropic axis is still incompletely understood. SerumGHBP levels appear to reflect GH responsivity ofthe organism, presumably by reflecting GHR statusin tissues. Regulation of serum GHBP levels is com-plex and in part variable among species; the principalregulators are ontogeny and development, nutri-tion, gender/estrogen, and in rodents pregnancy.Diagnostic use of the high affinity GHBP is currentlylimited to genetic GH insensitivity (Laron) syn-drome. GHBP can interfere in GH immunoassays,and GH assays need to be optimized in this regard.

The low affinity GHBP is transformed α2-macroglobulin. It appears to have limited import-ance for GH biology.

Acknowledgement

This chapter was supported in part by a merit re-view grant from the Department of VeteransAffairs.

References

Amit, T., Barkey, R.J., Youdim, M.B. &Hochberg, Z. (1990) A new andconvenient assay of growth hormone-binding protein activity in humanserum. Journal of Clinical Endocrinologyand Metabolism 71, 474–480.

Amit, T., Hertz, P., Ish-Shalom, S. et al.(1991) Effects of hypo or hyper-thyroidism on growth hormone-bindingprotein. Clinical Endocrinology 35,159–162.

Amit, T., Ish-Shalom, S., Glaser, B.,Youdim, M.B. & Hochberg, Z. (1992)Growth-hormone-binding protein inpatients with acromegaly. HormoneResearch 37, 205–211.

Amit, T., Dirnfeld, M., Barkey, R.J. et al.(1993) Growth hormone-binding protein (GHBP) levels in follicular fluidfrom human preovulatory follicles:correlation with serum GHBP levels.

Journal of Clinical Endocrinology andMetabolism 77, 33–39.

Ayling, R.M., Ross, R., Towner, P. et al.(1997) A dominant-negative mutation of the growth hormone receptor causesfamilial short stature. Nature Genetics 16,13–14.

Barnard, R., Quirk, P. & Waters, M.J. (1989) Characterization of the growthhormone-binding protein of humanserum using a panel of monoclonalantibodies. Journal of Endocrinology 123,327–332.

Barnard, R., Mulcahy, J., Garcia-Aragon, J.et al. (1994) Serum growth hormonebinding protein and hepatic GH bindingsites in the Lewis dwarf rat: effects ofIGF-I and GH. Growth Regulation 4,147–154.

Barsano, C.P. & Baumann, G. (1989) Simple algebraic and graphic methods

for the apportionment of hormone (andreceptor) into bound and free fractionsin binding equilibria; or how to calculatebound and free hormone? Endocrinology124, 1101–1106.

Baumann, G. (1995) Growth hormonebinding to a circulating receptorfragmentathe concept of receptorshedding and receptor splicing.Experimental and Clinical Endocrinology and Diabetes 103(1), 2–6.

Baumann, G. (2001) Growth hormonebinding protein 2001. Journal of PediatricEndocrinology and Metabolism 14,355–375.

Baumann, G. (2002) Geneticcharacterization of growth hormonedeficiency and resistance: implicationsfor treatment with recombinant growth hormone. American Journal ofPharmacogenomics 2, 93–111.

Page 121: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 105

Baumann, G. & Frank, S.J. (2002)Metalloproteinases and the modulationof growth hormone signalling. Journal ofEndocrinology 174, 361–368.

Baumann, G. & Shaw, M.A. (1988)Immunochemical similarity of thehuman plasma growth hormone-binding protein and the rabbit livergrowth hormone receptor. Biochemicaland Biophysical Research Communications152, 573–578.

Baumann, G. & Shaw, M.A. (1990) Asecond, lower affinity growth hormone-binding protein in human plasma.Journal of Clinical Endocrinology andMetabolism 70, 680–686.

Baumann, G., Stolar, M.W., Amburn, K.,Barsano, C.P. & DeVries, B.C. (1986) A specific growth hormone-bindingprotein in human plasma: initialcharacterization. Journal of ClinicalEndocrinology and Metabolism 62,134–141.

Baumann, G., Amburn, K.D. & Buchanan,T.A. (1987a) The effect of circulatinggrowth hormone-binding protein onmetabolic clearance, distribution, anddegradation of human growth hormone.Journal of Clinical Endocrinology andMetabolism 64, 657–660.

Baumann, G., Shaw, M.A. & Winter, R.J.(1987b) Absence of the plasma growthhormone-binding protein in Laron-type dwarfism. Journal of ClinicalEndocrinology and Metabolism 65,814–816.

Baumann, G., Amburn, K. & Shaw, M.A.(1988) The circulating growth hormone(GH)-binding protein complex: a majorconstituent of plasma GH in man.Endocrinology 122, 976–984.

Baumann, G., Shaw, M.A. & Amburn, K.(1989a) Regulation of plasma growthhormone-binding proteins in health anddisease. Metabolism 38, 683–689.

Baumann, G., Shaw, M.A. & Buchanan,T.A. (1989b) In vivo kinetics of a covalentgrowth hormone-binding proteincomplex. Metabolism 38, 330–333.

Baumann, G., Vance, M.L., Shaw, M.A. &Thorner, M.O. (1990) Plasma transportof human growth hormone in vivo.Journal of Clinical Endocrinology andMetabolism 71, 470–473.

Baumann, G., Lowman, H.B., Mercado, M.& Wells, J.A. (1994) The stoichiometry of growth hormone-binding proteincomplexes in human plasma:comparison with cell surface receptors.Journal of Clinical Endocrinology andMetabolism 78, 1113–1118.

protein-complex, and GH in healthychildren. Journal of Clinical Endocrinologyand Metabolism 77, 356–361.

Carmignac, D.F., Wells, T., Carlsson, L.M.,Clark, R.G. & Robinson, I.C. (1992)Growth hormone (GH) -binding proteinin normal and GH-deficient dwarf rats.Journal of Endocrinology 135, 447–457.

Carmignac, D.F., Gabrielsson, B.G. &Robinson, I.C. (1993) Growth hormonebinding protein in the rat: effects ofgonadal steroids. Endocrinology 133,2445–2452.

Cerio, R.J., Xing, F., Fatula, R.J. et al. (2002) Structurally distinct membrane-associated and soluble forms of GH-binding protein in the mouse. Journal ofEndocrinology 172, 321–331.

Chapman, I.M., Hartman, M.L., Straume, M. et al. (1994) Enhancedsensitivity growth hormone (GH)chemiluminescence assay reveals lowerpostglucose nadir GH concentrations in men than women. Journal of ClinicalEndocrinology and Metabolism 78,1312–1319.

Clark, R.G., Mortensen, D.L., Carlsson,L.M. et al. (1996) Recombinant humangrowth hormone (GH)-binding proteinenhances the growth-promoting activityof human GH in the rat. Endocrinology137, 4308–4315.

Collipp, P.J., Kaplan, S.A., Boyle, D.C. &Shimizu, C.S.N. (1964) Protein-boundhuman growth hormone. Metabolism 13,532–538.

Cramer, S.D., Barnard, R., Engbers, C.,Thordarson, G. & Talamantes, F. (1992)A mouse growth hormone-bindingprotein RIA: concentrations in maternalserum during pregnancy. Endocrinology130, 1074–1076.

Dastot, F., Sobrier, M.L., Duquesnoy, P. et al. (1996) Alternatively spliced formsin the cytoplasmic domain of the humangrowth hormone (GH) receptor regulateits ability to generate a soluble GH-binding protein. Proceedings of theNational Academy of Sciences of the UnitedStates of America 93, 10 723–10 728.

Daughaday, W.H. & Trivedi, B. (1987)Absence of serum growth hormonebinding protein in patients with growthhormone receptor deficiency (Larondwarfism). Proceedings of the NationalAcademy of Sciences of the United States ofAmerica 84, 4636–4640.

Daughaday, W.H., Trivedi, B. & Andrews,B.A. (1987) The ontogeny of serum GHbinding protein in man: a possibleindicator of hepatic GH receptor

Baumbach, W.R., Horner, D.L. & Logan,J.S. (1989) The growth hormone-bindingprotein in rat serum is an alternativelyspliced form of the rat growth hormonereceptor. Genes and Development 3,1199–1205.

Baxter, R.C. & Turtle, J.R. (1978)Regulation of hepatic growth hormonereceptors by insulin. Biochemical andBiophysical Research Communications 84,350–357.

Berson, S.A. & Yalow, R.S. (1966a) Peptidehormones in plasma. Harvey Lectures 62,107–163.

Berson, S.A. & Yalow, R.S. (1966b) State ofhuman growth hormone in plasma andchanges in stored solutions of pituitarygrowth hormone. Journal of BiologicalChemistry 241, 5745–5749.

Black, R.A., Rauch, C.T., Kozlosky, C.J. et al. (1997) A metalloproteinasedisintegrin that releases tumour-necrosis factor-α from cells. Nature 385,729–733.

Blumenfeld, Z., Barkey, R.J., Youdim, M.B., Brandes, J.M. & Amit, T. (1992)Growth hormone (GH) -binding protein regulation by estrogen,progesterone, and gonadotropins inhuman: the effect of ovulation inductionwith menopausal gonadotropins, GH,and gestation. Journal of ClinicalEndocrinology and Metabolism 75,1242–1249.

Camarillo, I.G., Thordarson, G., Ilkbahar,Y.N. & Talamantes, F. (1998)Development of a homologousradioimmunoassay for mouse growthhormone receptor. Endocrinology 139,3585–3589.

Carlsson, B., Billig, H., Rymo, L. &Isaksson, O.G. (1990) Expression of the growth hormone-binding proteinmessenger RNA in the liver andextrahepatic tissues in the rat: co-expression with the growth hormonereceptor. Molecular and CellularEndocrinology 73, R1–R6.

Carlsson, L.M., Rowland, A.M., Clark, R.G., Gesundheit, N. & Wong,W.L. (1991) Ligand-mediatedimmunofunctional assay forquantitation of growth hormone-binding protein in human blood. Journalof Clinical Endocrinology and Metabolism73, 1216–1223.

Carlsson, L.M., Rosberg, S., Vitangcol,R.V., Wong, W.L. & Albertsson-Wikland, K. (1993) Analysis of 24-hourplasma profiles of growth hormone(GH)-binding protein, GH/GH-binding

Page 122: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

106 chapter 8

development. Journal of ClinicalEndocrinology and Metabolism 65,1072–1074.

Duquesnoy, P., Sobrier, M.L., Duriez, B. et al. (1994) A single amino acidsubstitution in the exoplasmic domain of the human growth hormone (GH)receptor confers familial GH resistance(Laron syndrome) with positive GH-binding activity by abolishing receptorhomodimerization. EMBO Journal 13,1386–1395.

Edens, A., Southard, J.N. & Talamantes, F.(1994) Mouse growth hormone-bindingprotein and growth hormone receptortranscripts are produced from a singlegene by alternative splicing.Endocrinology 135, 2802–2805.

Eliakim, A., Brasel, J.A., Barstow, T.J.,Mohan, S. & Cooper, D.M. (1998a) Peakoxygen uptake, muscle volume, and thegrowth hormone-insulin-like growthfactor-I axis in adolescent males.Medicine and Science in Sports and Exercise30, 512–517.

Eliakim, A., Brasel, J.A., Mohan, S., Wong,W.L. & Cooper, D.M. (1998b) Increasedphysical activity and the growthhormone-IGF-I axis in adolescent males.American Journal of Physiology 275,R308–R314.

Eliakim, A., Scheett, T.P., Newcomb, R.,Mohan, S. & Cooper, D.M. (2001)Fitness, training, and the growthhormone-insulin-like growth factor Iaxis in prepubertal girls. Journal ofClinical Endocrinology and Metabolism 86,2797–2802.

Fisker, S., Frystyk, J., Skriver, L. et al. (1996)A simple, rapid immunometric assay fordetermination of functional and growthhormone-occupied growth hormone-binding protein in human serum.European Journal of Clinical Investigation26, 779–785.

Fisker, S., Ebdrup, L. & Orskov, H. (1998)Influence of growth hormone bindingprotein on growth hormone estimationin different immunoassays. ScandinavianJournal of Clinical and LaboratoryInvestigation 58, 373–381.

Forbes, G.B. (1977) Nutrition and growth.Journal of Pediatrics 91, 40–42.

Frick, G.P., Tai, L.R. & Goodman, H.M.(1994) Subcellular distribution of thelong and short isoforms of the growthhormone (GH) receptor in ratadipocytes: both isoforms participate inspecific binding of GH. Endocrinology134, 307–314.

Frick, G.P., Tai, L.R., Baumbach, W.R. & Goodman, H.M. (1998) Tissue

Herington, A.C., Ymer, S. & Stevenson, J.(1986b) Identification andcharacterization of specific bindingproteins for growth hormone in normalhuman sera. Journal of ClinicalInvestigation 77, 1817–1823.

Ho, K.K., Valiontis, E., Waters, M.J. &Rajkovic, I.A. (1993) Regulation ofgrowth hormone binding protein inman: comparison of gel chromatographyand immunoprecipitation methods.Journal of Clinical Endocrinology andMetabolism 76, 302–308.

Hochberg, Z., Hertz, P., Colin, V. et al.(1992) The distal axis of growth hormone(GH) in nutritional disorders: GH-binding protein, insulin-like growthfactor-I (IGF-I), and IGF-I receptors inobesity and anorexia nervosa.Metabolism 41, 106–112.

Holl, R.W., Snehotta, R., Siegler, B.,Scherbaum, W. & Heinze, E. (1991)Binding protein for human growthhormone: effects of age and weight.Hormone Research 35, 190–197.

Iida, K., Takahashi, Y., Kaji, H. et al. (1998) Growth hormone (GH)insensitivity syndrome with high serumGH-binding protein levels caused by aheterozygous splice site mutation of theGH receptor gene producing a lack ofintracellular domain. Journal of ClinicalEndocrinology and Metabolism 83,531–537.

Iida, K., Takahashi, Y., Kaji, H. et al. (1999)Functional characterization of truncatedgrowth hormone (GH) receptor-(1-277)causing partial GH insensitivitysyndrome with high GH-bindingprotein. Journal of Clinical Endocrinologyand Metabolism 84, 1011–1016.

Irie, M. & Barrett, R.J. (1962) Immunologicstudies of human growth hormone.Endocrinology 71, 277–287.

Jan, T., Shaw, M.A. & Baumann, G. (1991)Effects of growth hormone-bindingproteins on serum growth hormonemeasurements. Journal of ClinicalEndocrinology and Metabolism 72,387–391.

Jansson, C., Boguszewski, C., Rosberg, S.,Carlsson, L.M. & Albertsson-Wikland,K. (1997) Growth hormone (GH) assays:influence of standard preparations, GHisoforms, assay characteristics, and GH-binding protein. Clinical Chemistry 43,950–956.

Kaji, H., Nose, O., Tajiri, H. et al. (1997)Novel compound heterozygousmutations of growth hormone (GH)receptor gene in a patient with GHinsensitivity syndrome. Journal of Clinical

distribution, turnover, and glycosylationof the long and short growth hormonereceptor isoforms in rat tissues.Endocrinology 139, 2824–2830.

Gabrielsson, B.G., Carmignac, D.F., Flavell,D.M. & Robinson, I.C. (1995) Steroidregulation of growth hormone (GH)receptor and GH-binding proteinmessenger ribonucleic acids in the rat.Endocrinology 136, 209–217.

Gastier, J.M., Berg, M.A., Vesterhus, P.,Reiter, E.O. & Francke, U. (2000) Diversedeletions in the growth hormonereceptor gene cause growth hormoneinsensitivity syndrome. Human Mutation16, 323–333.

Gelander, L., Bjarnason, R., Carlsson, L.M. & Albertsson-Wikland, K. (1998)Growth hormone-binding protein levelsover 1 year in healthy prepubertalchildren: intraindividual variation andcorrelation with height velocity. PediatricResearch 43, 256–261.

Hadden, D.R. & Prout, T.E. (1964) Agrowth hormone binding protein innormal human serum. Nature 202,1342–1343.

Hanaire-Broutin, H., Sallerin-Caute, B.,Poncet, M.F. et al. (1996) Effect ofintraperitoneal insulin delivery ongrowth hormone binding protein,insulin-like growth factor (IGF) -I, andIGF-binding protein-3 in IDDM.Diabetologia 39, 1498–1504.

Harada, I., Tsutsumi, O., Momoeda, M. et al. (1997) Comparative concentrationsof growth hormone-binding protein inmaternal circulation, fetal circulation,and amniotic fluid. Endocrine Journal 44,111–116.

Hattori, N., Shimatsu, A., Kato, Y. &Imura, H. (1990) Growth hormone andgrowth hormone binding protein inhuman urine. Kidney International 37,951–954.

Hattori, N., Kurahachi, H., Ikekubo, K. et al. (1991) Effects of sex and age onserum GH binding protein levels innormal adults. Clinical Endocrinology 35,295–297.

Heinrichs, C., Yanovski, J.A., Roth, A.H. et al. (1994) Dexamethasone increasesgrowth hormone receptor messengerribonucleic acid levels in liver andgrowth plate. Endocrinology 135,1113–1118.

Herington, A.C., Ymer, S., Roupas, P. &Stevenson, J. (1986a) Growth hormone-binding proteins in high-speed cytosolsof multiple tissues of the rabbit.Biochimica et Biophysica Acta 881,236–240.

Page 123: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 107

Endocrinology and Metabolism 82,3705–3709.

Keenan, B.S., Richards, G.E., Mercado, M.et al. (1996) Androgen regulation ofgrowth hormone binding protein.Metabolism 45, 1521–1526.

Kratzsch, J., Blum, W.F., Ventz, M. et al.(1995a) Growth hormone-bindingprotein-related immunoreactivity in theserum of patients with acromegaly isregulated inversely by growth hormoneconcentration. European Journal ofEndocrinology 132, 306–312.

Kratzsch, J., Selisko, T. & Birkenmeier, G.(1995b) Identification of transformed α2-macroglobulin as a growth hormone-binding protein in human blood. Journalof Clinical Endocrinology and Metabolism80, 585–590.

Kratzsch, J., Keliner, K., Zilkens, T. et al.(1996) Growth hormone-binding proteinrelated immunoreactivity is regulatedby the degree of insulinopenia indiabetes mellitus. Clinical Endocrinology44, 673–678.

Kratzsch, J., Dehmel, B., Pulzer, F. et al.(1997a) Increased serum GHBP levels inobese pubertal children and adolescents:relationship to body composition, leptin and indicators of metabolicdisturbances. International Journal ofObsesity and Related Metabolic Disorders21, 1130–1136.

Kratzsch, J., Schreiber, G., Selisko, T. et al.(1997b) Measurement of serum exon 3-retaining growth hormone-bindingprotein in children and adolescents byradioimmunoassay. Hormone Research48, 252–257.

Leung, D.W., Spencer, S.A., Cachianes, G.et al. (1987) Growth hormone receptorand serum binding protein: purification,cloning and expression. Nature 330,537–543.

Leung, K.C., Doyle, N. & Ho, K.K. (2000)Characterization of a low affinitybinding protein for growth hormone inrat serum. Endocrinology 141, 138–145.

Lim, L., Spencer, S.A., McKay, P. & Waters,M.J. (1990) Regulation of growthhormone (GH) bioactivity by arecombinant human GH-bindingprotein. Endocrinology 127, 1287–1291.

Lobie, P.E., Barnard, R. & Waters, M.J.(1991) The nuclear growth hormonereceptor binding protein. Antigenic and physicochemical characterization.Journal of Biological Chemistry 266, 22 645–22 652.

Lobie, P.E., Garcia-Aragon, J., Wang, B.S.,Baumbach, W.R. & Waters, M.J. (1992)Cellular localization of the growth

Mercado, M. & Baumann, G. (1994) Agrowth hormone/prolactin-bindingprotein in human milk. Journal of ClinicalEndocrinology and Metabolism 79,1637–1641.

Mercado, M., Molitch, M.E. & Baumann, G. (1992) Low plasma growth hormonebinding protein in IDDM. Diabetes 41,605–609.

Mercado, M., Carlsson, L.M., Vitangcol, R. & Baumann, G. (1993) Growthhormone-binding protein determinationin plasma: a comparison ofimmunofunctional and growthhormone-binding assays. Journal ofClinical Endocrinology and Metabolism 76,1291–1294.

Miell, J.P., Buchanan, C.R., Norman, M.R., Maheshwari, H.G. & Blum, W.F.(1994) The evolution of changes inimmunoreactive serum insulin-likegrowth factors (IGFs), IGF-bindingproteins, circulating growth hormone(GH) and GH-binding protein as a result of short-term dexamethasonetreatment. Journal of Endocrinology 142,547–554.

Mulumba, N., Massa, G., Ketelslegers, J.M.& Maes, M. (1991) Ontogeny andnutritional regulation of the serumgrowth hormone-binding protein in therat. Acta Endocrinologica 125, 409–415.

Nixon, D.A. & Jordan, R.M. (1986)Conversion of CSF monomeric growthhormone to large growth hormone withexposure to serum. Acta Endocrinologica111, 289–295.

Pantel, J., Machinis, K., Sobrier, M.L. et al.(2000) Species-specific alternative splicemimicry at the growth hormonereceptor locus revealed by the lineage ofretroelements during primate evolution.Journal of Biological Chemistry 275, 18 664–18 669.

Peeters, S. & Friesen, H.G. (1977) A growthhormone binding factor in the serum of pregnant mice. Endocrinology 101,1164–1183.

Postel-Vinay, M.C., Cohen-Tanugi, E. &Charrier, J. (1982) Growth hormonereceptors in rat liver membranes: effectsof fasting and refeeding, and correlationwith plasma somatomedin activity.Molecular and Cellular Endocrinology 28,657–669.

Postel-Vinay, M.C., Belair, L., Kayser, C.,Kelly, P.A. & Djiane, J. (1991a)Identification of prolactin and growthhormone binding proteins in rabbitmilk. Proceedings of the National Academyof Sciences of the United States of America88, 6687–6690.

hormone binding protein in the rat.Endocrinology 130, 3057–3065.

Maheshwari, H., Lillioja, S., Castillo, C.E.,Mercado, M. & Baumann, G. (1995)Growth hormone-binding protein inhuman lymph. Journal of ClinicalEndocrinology and Metabolism 80,3582–3584.

Maheshwari, H., Sharma, L. & Baumann,G. (1996) Decline of plasma growthhormone binding protein in old age.Journal of Clinical Endocrinology andMetabolism 81, 995–997.

Mandell, F. & Berenberg, W. (1974) TheMauriac syndrome. American Journal ofDiseases of Children (1960) 127, 900–902.

Mannor, D.A., Winer, L.M., Shaw, M.A. &Baumann, G. (1991) Plasma growthhormone (GH)-binding proteins: effecton GH binding to receptors and GHaction. Journal of Clinical Endocrinologyand Metabolism 73, 30–34.

Martha, P.M., Jr., Reiter, E.O., Davila, N. et al. (1992) The role of body mass in theresponse to growth hormone therapy.Journal of Clinical Endocrinology andMetabolism 75, 1470–1473.

Martha, P.M., Jr., Rogol, A.D., Carlsson,L.M., Gesundheit, N. & Blizzard, R.M.(1993) A longitudinal assessment ofhormonal and physical alterationsduring normal puberty in boys. I. Serumgrowth hormone-binding protein.Journal of Clinical Endocrinology andMetabolism 77, 452–457.

Martini, J.F., Pezet, A., Guezennec, C.Y. et al. (1997) Monkey growth hormone(GH) receptor gene expression.Evidence for two mechanisms for thegeneration of the GH binding protein.Journal of Biological Chemistry 272, 18 951–18 958.

Massa, G., Mulumba, N., Ketelslegers, J.M. & Maes, M. (1990) Initialcharacterization and sexual dimorphismof serum growth hormone-bindingprotein in adult rats. Endocrinology 126,1976–1980.

Massa, G., Verhaeghe, J., Vanderschueren-Lodeweyckx, M. & Bouillon, R. (1993)Normalization of decreased plasmaconcentrations of growth hormone-binding protein by insulin treatment inspontaneously diabetic BB rats. Hormoneand Metabolic Research 25, 325–326.

Mauras, N., Merimee, T. & Rogol, A.D.(1991) Function of the growth hormone-insulin-like growth factor I axis in theprofoundly growth-retarded diabeticchild: evidence for defective target organ responsiveness in the Mauriacsyndrome. Metabolism 40, 1106–1111.

Page 124: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

108 chapter 8

Postel-Vinay, M.C., Tar, A., Hocquette, J.F.et al. (1991b) Human plasma growthhormone (GH)-binding proteins areregulated by GH and testosterone.Journal of Clinical Endocrinology andMetabolism 73, 197–202.

Rajkovic, I.A., Valiontis, E. & Ho, K.K.(1994) Direct quantitation of growthhormone binding protein in humanserum by a ligand immunofunctionalassay: comparison withimmunoprecipitation andchromatographic methods. Journal ofClinical Endocrinology and Metabolism 78,772–777.

Roelen, C.A., Donker, G.H., Thijssen, J.H.,Koppeschaar, H.P. & Blankenstein, M.A.(1992) High affinity growth hormonebinding protein in plasma of patientswith acromegaly and the effect ofoctreotide treatment. ClinicalEndocrinology 37, 373–378.

Roelen, C.A., de Vries, W.R., Koppeschaar,H.P. et al. (1997a) Plasma insulin-likegrowth factor-I and high affinity growthhormone-binding protein levels increaseafter 2 weeks of strenuous physicaltraining. International Journal of SportsMedicine 18, 238–241.

Roelen, C.A., Koppeschaar, H.P., de Vries,W.R. et al. (1997b) Visceral adipose tissueis associated with circulating highaffinity growth hormone-bindingprotein. Journal of Clinical Endocrinologyand Metabolism 82, 760–764.

Roemmich, J.N. & Sinning, W.E. (1997)Weight loss and wrestling training:effects on growth-related hormones.Journal of Applied Physiology 82,1760–1764.

Romero, G.S., Stephan, D.A., Sperling,M.A. & Menon, R.K. (1996) Distinctsexual dimorphism in the effect ofhypothyroidism on the expression of thegrowth hormone receptor and growthhormone-binding protein gene in ratliver. Hormone Research 45, 273–278.

Rosenfeld, R.G., Rosenbloom, A.L. &Guevara-Aguirre, J. (1994) Growthhormone (GH) insensitivity due toprimary GH receptor deficiency.Endocrine Reviews 15, 369–390.

Ross, R.J., Esposito, N., Shen, X.Y. et al.(1997) A short isoform of the humangrowth hormone receptor functions as adominant negative inhibitor of the full-length receptor and generates largeamounts of binding protein. MolecularEndocrinology 11, 265–273.

Ross, R.J., Leung, K.C., Maamra, M. et al.(2001) Binding and functional studieswith the growth hormone receptor

Endocrinology and Metabolism 70,417–420.

Spencer, S.A., Hammonds, R.G., Henzel,W.J. et al. (1988) Rabbit liver growthhormone receptor and serum bindingprotein. Purification, characterization,and sequence. Journal of BiologicalChemistry 263, 7862–7867.

Tar, A., Hocquette, J.F., Souberbielle, J.C. et al. (1990) Evaluation of the growthhormone-binding proteins in humanplasma using high pressure liquidchromatography gel filtration. Journal ofClinical Endocrinology and Metabolism 71,1202–1207.

Touber, J.L. & Maingay, D. (1963)Heterogeneity of human growthhormone. Its influence on a radio-immunoassay of the hormone in serum.Lancet 1, 403–405.

Veldhuis, J.D., Johnson, M.L., Faunt, L.M.,Mercado, M. & Baumann, G. (1993)Influence of the high-affinity growthhormone (GH) -binding protein onplasma profiles of free and bound GHand on the apparent half-life of GH.Modeling analysis and clinicalapplications. Journal of ClinicalInvestigation 91, 629–641.

Walker, J.L., Moats-Staats, B.M., Stiles,A.D. & Underwood, L.E. (1992) Tissue-specific developmental regulation of themessenger ribonucleic acids encodingthe growth hormone receptor and thegrowth hormone binding protein in ratfetal and postnatal tissues. PediatricResearch 31, 335–339.

Wallace, J.D., Cuneo, R.C., Baxter, R. et al. (1999) Responses of the growthhormone (GH) and insulin-like growth factor axis to exercise, GHadministration, and GH withdrawal intrained adult males: a potential test forGH abuse in sport. Journal of ClinicalEndocrinology and Metabolism 84,3591–3601.

Wang, X., He, K., Gerhart, M. et al. (2002) Metalloprotease-mediated GH receptor proteolysis and GHBPshedding. Determination of extracellulardomain stem region cleavage site.Journal of Biological Chemistry 277, 50 510–50 519.

Weissberger, A.J., Ho, K.K. & Lazarus, L.(1991) Contrasting effects of oral andtransdermal routes of estrogenreplacement therapy on 24-hour growthhormone (GH) secretion, insulin-likegrowth factor I, and GH-binding proteinin postmenopausal women. Journal ofClinical Endocrinology and Metabolism 72,374–381.

antagonist, B2036-PEG (pegvisomant),reveal effects of pegylation and evidencethat it binds to a receptor dimer. Journalof Clinical Endocrinology and Metabolism86, 1716–1723.

Sadeghi, H., Wang, B.S., Lumanglas, A.L.,Logan, J.S. & Baumbach, W.R. (1990)Identification of the origin of the growthhormone-binding protein in rat serum.Molecular Endocrinology 4, 1799–1805.

Sanchez-Jimenez, F., Fielder, P.J.,Martinez, R.R., Smith, W.C. &Talamantes, F. (1990) Hypophysectomyeliminates and growth hormone (GH)maintains the midpregnancy elevationin GH receptor and serum bindingprotein in the mouse. Endocrinology 126,1270–1275.

Scheett, T.P., Nemet, D., Stoppani, J. et al.(2002) The effect of endurance-typeexercise training on growth mediatorsand inflammatory cytokines in pre-pubertal and early pubertal males.Pediatric Research 52, 491–497.

Seidel, B., Glasow, A., Schutt, M. et al.(2003) Association between the GHreceptor/exon 3 genotype and the levelof exon 3-positive GH-binding protein in human serum. European Journal ofEndocrinology 148, 317–324.

Silbergeld, A., Klinger, B., Keret, R. et al.(1994) Serum growth hormone-bindingprotein (GHBP) activity is decreased byadministration of insulin-like growthfactor I in three Laron syndrome siblingswith normal GHBP. Proceeding of theSociety for Experimental Biology andMedicine 206, 324–327.

Silbergeld, A., Dastot, F., Klinger, B. et al.(1997) Intronic mutation in the growthhormone (GH) receptor gene from a girlwith Laron syndrome and extremelyhigh serum GH binding protein:extended phenotypic study in a verylarge pedigree. Journal of PediatricEndocrinology and Metabolism 10,265–274.

Smith, W.C. & Talamantes, F. (1988)Gestational profile and affinity cross-linking of the mouse serum growthhormone-binding protein. Endocrinology123, 1489–1494.

Smith, W.C., Kuniyoshi, J. & Talamantes,F. (1989) Mouse serum growth hormone(GH) binding protein has GH receptorextracellular and substitutedtransmembrane domains. MolecularEndocrinology (Baltimore, Md) 3, 984–990.

Snow, K.J., Shaw, M.A., Winer, L.M. &Baumann, G. (1990) Diurnal pattern ofplasma growth hormone-bindingprotein in man. Journal of Clinical

Page 125: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone binding proteins 109

Woods, K.A., Fraser, N.C., Postel-Vinay,M.C., Savage, M.O. & Clark, A.0J. (1996)A homozygous splice site mutationaffecting the intracellular domain of the growth hormone (GH) receptorresulting in Laron syndrome withelevated GH-binding protein. Journal ofClinical Endocrinology and Metabolism 81,1686–1690.

Woods, K.A., Dastot, F., Preece, M.A. et al. (1997) Phenotype: genotyperelationships in growth hormoneinsensitivity syndrome. Journal of ClinicalEndocrinology and Metabolism 82,3529–3535.

Ymer, S.I. & Herington, A.C. (1985)Evidence for the specific binding ofgrowth hormone to a receptor-like

Zhang, Y., Guan, R., Jiang, J. et al. (2001)Growth hormone (GH)-induceddimerization inhibits phorbol ester-stimulated GH receptor proteolysis.Journal of Biological Chemistry 276, 24 565–24 573.

Zhou, Y., He, L. & Kopchick, J.J. (1994) An exon encoding the mouse growthhormone binding protein (mGHBP)carboxy terminus is located betweenexon 7 and 8 of the mouse growthhormone receptor gene. Receptor 4,223–227.

Zhou, Y., He, L. & Kopchick, J.J. (1996)Structural comparison of a portion of the rat and mouse growth hormonereceptor/binding protein genes. Gene177, 257–259.

protein in rabbit serum. Molecular andCellular Endocrinology 41, 153–161.

Yu, Y.M., Domene, H.M., Sztein, J., Counts, D.R. & Cassorla, F. (1996)Developmental changes and differentialregulation by testosterone and estradiolof growth hormone receptor expressionin the rabbit. European Journal ofEndocrinology 135, 583–590.

Zhang, Y., Jiang, J., Black, R.A., Baumann, G. & Frank, S.J. (2000) TACE is a growth hormone bindingprotein (GHBP) sheddase: themetalloprotease TACE/ADAM-17 iscritical for (PMA-induced) growthhormone receptor proteolysis and GHBP generation. Endocrinology 141,4342–4348.

Page 126: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

110

Introduction

Resistance exercise is the most prolific form of exercise producing anabolic effects in muscle andconnective tissues (Kraemer et al. 1996). Humangrowth hormone (GH), also called somatotropin, a pleiotropic polypeptide, exerts a multitude ofeffects upon the metabolic state of the human body,in part mediating a myriad of metabolic and growthprocesses. The complexity of pituitary release andproduction of GH, aggregates, and binding proteinsis just beginning to be elucidated with the inter-pretation of the literature on resistance exercise andtraining remaining a puzzle with parts of the picturestill requiring study (Nindl et al. 2003). Neverthe-less, understanding the responses and adaptationsof the 22 kDa GH will continue to play an importantpart of our understanding of the adaptations toresistance training. The hGH-N gene expresses themain pituitary molecular weight variant in the GHfamily which is the 22 kDa form. This protein is 191amino acids in length (monomeric 22 kDa repres-ents ~ 21% of all circulating plasma GH). The nextmost prevalent form is the 20 kDa molecule (mono-meric 20 kDa represents ~ 6% of all circulatingplasma GH) formed through alternative mRNAsplicing during which amino acid residues 32–46are cleaved out. Human GH can also undergo post-translational modification and peripheral tissueproteolytic cleavage at its site of action to form variants and aggregates (i.e. dimers, trimers, penta-mers) and fragments which exist in the circulation.The existence of high- and low-affinity GH bind-ing proteins (released from the pituitary and also

cleaved from the extracellular domain of the GHreceptor) adds further complexity to the nature andspatial arrangement of circulating GH moieties.Thus, within the limitations for the scope of thischapter we will examine some of the basic responsesand adaptations of the 22 kDa GH to resistance exercise and training.

Growth hormone release and control

Many pituitary hormones are under hypothalamiccontrol. The hypothalamic hormones (e.g. releas-ing factors) growth hormone releasing hormone(GHRH) and somatostatin (SS) serve to stimulateand inhibit GH release, respectively. These releas-ing factors are secreted from neurons originating inthe arcuate, periventricular and paraventricularnuclei (i.e. hypothalamic nuclei) whose axon ter-minals extend toward the median eminence. Thereleasing factors are delivered into a portal systemof veins which acts as a humoral pathway for directdelivery to the anterior pituitary. The portal bloodsystem originates from capillary loops in the medianeminence of the tuber cinereum and blood drains ina parallel fashion down the pituitary stalk. In theanterior pituitary, the portal blood vessels break up into the sinusoids which provide the nutrientsupply. This vascular supply is very small and thecapillary permeability is quite high. In this manner,releasing factors from the hypothalamus may reachsecreting cells (i.e. somatotrophs) of the anteriorpituitary (Fig. 9.1). Functionally distinct types (i.e.type I and II or band 1 and band II cells) of somato-trophs have been identified using density gradient

Chapter 9

Resistance Exercise: Acute and Chronic Changes inGrowth Hormone Concentrations

WILLIAM J. KRAEMER, BRADLEY C. NINDL AND SCOTT E. GORDON

Page 127: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 111

centrifugation. The somatotrophs in these two fractions can be distinguished morphologically bytheir staining characteristics and ultrastructure, andhave been shown to vary in their responsiveness to GH secreting stimuli with the type I being moreresponsive (Snyder et al. 1977). The physiologicalsignificance of this cellular heterogeneity andwhether exercise can differentially influence thesecells remains unknown, but presents a provocativearea of future study as the complexities of exercise-induced release and adaptations interact with theheterogeneity of the different molecular weightforms of GH and GH aggregates.

The central mechanisms underlying pulsatile GHrelease are classically thought to involve elevatedsecretion of GHRH into the hypophysial portalblood during troughs or nadirs of SS secretion. Theprecise molecular mechanisms responsible for thispulsatility remain speculative. Superimposed onthis basic mechanism are other control and modu-latory mechanisms that determine somatotrophresponsiveness at any give point in time. There isevidence that GHRH affects both GH biosynthesisand release, and SS inhibits release without affect-ing biosynthesis. It is also thought that GHRH isrequired for the initiation of GH pulses, while SSdictates the amplitude of the pulse. SS control over

GH pulse amplitude is supported by studies thathave reported that the GH response to GHRH isenhanced by administration of SS antagonists (viz.pyridostigmine [an acetylcholinesterase inhibitor]and hexapeptide GH-releasing peptides) (Cappa et al.1993). The cloning of a GH secretagogue receptorhas also demonstrated that the GH secretagogue, thepreviously elusive endogenous ligand of the recep-tor (recently identified as ghrelin), is part of a newphysiological regulating system in GH secretion.

It has been well established that GH is secretedfrom the anterior pituitary gland in an episodic, pulsatile manner throughout the day with a dramaticsurge of release during slow-wave sleep (Fig. 9.2).This pulsatile release is under the regulatory controlof the two hypothalamic hormones that serve tostimulate (i.e. growth factor releasing hormone) orinhibit (i.e. SS) GH release. The balance betweenthese two hormones determines the relative magni-tude of initial release from the anterior pituitary.

It does appear that GH can exert acute negativefeedback on its own release. When subjects areadministered a single dose of GH, subsequent GHresponses to GHRH are diminished or abolished(Scanlon et al. 1996). This ‘somatotroph desensitiza-tion’ mechanism can be reversed by prior activationof cholinergic pathways. Currently, it appears that

Posterior pituitary

Median eminence

Release ofhypothalamic

releasing factors

Arcuate nucleiParaventricular nuclei

Supraoptic nucleusHypothalamic neurons

Hypothalamus

Stalk

Portal vessels

Secreting cell ofanterior pituitary(Somatotroph)

Anterior pituitary

Fig. 9.1 Hypothalamic–pituitaryneural interface regulating growthhormone (GH) secretion.

Page 128: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

112 chapter 9

alterations in GH release are mediated through aninhibition of SS secreting cells via cholinergic path-ways (Giustina & Veldhuis 1998). As noted above SSinhibits GH release, but not its synthesis. This is animportant concept as it may explain ‘rebound’ GHsecretion after SS priming and withdrawal (Giustina& Veldhuis 1998).

There are many neuromodulators (i.e. neuropep-tides, neurotransmitters, physiological conditions)that have putative roles in regulating GH secretionvia GHRH and SS have been an intense area of regu-latory mechanism research. Veldhuis et al. (2004)have postulated ‘that sex-steroid-specific control of SS and GHRH outflow may mediate the for-mer gender contrasts, whereas unknown (gender-independent) factors may determine the capabilityof exercise to significantly antagonize GH auto-inhibition’. Table 9.1 lists a summary of theseknown modulators. These factors act to stimulate(α1-adrenergic, amino acids, dopamine, muscariniccholinergic, GABA(-B), galanin, growth hormonereleasing peptide (GHRP), histamine, hypoglycemia,neuromedin C, opiates, serotonin, diabetes, acuteand chronic exercise, starvation and stress) or inhi-bit (α1-adrenergic, GHRH immunization cortisol/glucocorticoids, glucose, hypothyroidism, obesity,aging) GH release. Peripheral feedback regulationof the somatotroph is mediated by the array of GHtarget actions. Namely, insulin-like growth factor I(IGF-I), glucose and free fatty acids can each exertfeedback influences at hypothalamic/pituitary lev-els. The array of multivariate neuroregulatory fac-tors shown in Table 9.1 that influence GH releaseemphasize the concept that GH release is a complex

issue. The interplay between exercise and many ofthese factors in the control of GH release is not fullyunderstood.

Growth hormone release patterns

Two unique characteristics of GH are its pulsatilerelease and the degree of molecular heterogeneity.The GH-N gene expresses the main pituitary mole-cular weight variant in the GH family, which is the22 kDa form. This protein is 191 amino acids inlength with two disulfide cross-linkages (mono-meric 22 kDa represents ~ 21% of all circulatingplasma GH). The next most prevalent form is the 20 kDa molecule (monomeric 20 kDa represents ~ 6% of all circulating plasma GH) formed throughalternative mRNA splicing during which aminoacid residues 32–46 are cleaved out. Human GH canalso undergo post-translational modification andperipheral tissue proteolytic cleavage at its site ofaction to form variants and aggregates (i.e. dimers,trimers, pentamers) and fragments which exist inthe circulation. The existence of high- and low-affinity GH binding proteins (released from thepituitary and also cleaved from the extracellulardomain of the GH receptor) adds further complex-ity to the nature and spatial arrangement of circulat-ing GH moieties. This molecular heterogeneityappears to have physiological significance as thedifferent forms have been shown to possess dif-ferent biological activities (e.g. relative potency inbioassays) as well as their ability to be detected inimmunoassays. The contents of the band I and bandII secretory cells in the pituitary can impact the

02468

1012141618202224

1700

1900

2100

2300 100

300

500

Gro

wth

ho

rmo

ne

(ng

·mL–1

)

Time of day (h)

Fig. 9.2 Example 13-h growthhormone (GH) pulsatility profile for a young health male. Note that the greatest pulse amplitudeoccurred during sleeping hours(approximately 0200 h).

Page 129: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 113

profile of the composition of the circulatory concen-trations of various GH isoforms and may eventuallyprovide the insights into the vast array of physiolo-gical mechanisms related to GH, yet not explainedby the 22 kDa molecule alone (Hymer et al. 2001).

Growth hormone secretion

Many factors affecting GH secretion including age,gender, diet and nutrients, stress, other hormones

(e.g. gonadal steroids, thyroid hormones and IGF-I),adiposity, fitness level and exercise have all beenimplicated as factors that influence GH concentra-tions in the blood (see Fig. 9.1). Since the initialobservations of Hunter et al. (1965), it is well recog-nized that physical activity is a naturally occurringstimulator of GH release into the circulation. GHhas been linked to the promotion of anabolism inboth muscle and connective tissue. Specifically, itenhances cellular amino acid uptake and protein

Table 9.1 Neuroregulatory modulators of growth hormone (GH) secretion in humans.

Effector Effect

Adrenergic (α1, α2 & β2) No effect, ↑, ↓Age ↓Amino acids ↑Autofeedback at hypothalamus by IGF-I ↓Bombesin No effect basally, ↓ hypoglycemia effectDopamine ↑Muscarinic (chlolinergic & nicotinic) ↑ & ↓Cortisol/glucocorticoids ↓Diabetes mellitus (type 1 & type 2) ↑ & ↑ or ↓Estrogen ↑ AmplitudeExcitatory amino acids UnknownExercise (acute & chronic) ↑ & ↑Fatty acids ↓GABA(-B) ↑ Basal ↓ (stim)Galanin ↑GHRP ↑Glucose ↓Histamine ↑Hypoglycemia ↓Hypothryrodisim ↓IGF-I (pituitary inhibition) YesImmunization (or antagonist) (GHRH & SS) ↓ Amplitude & unknownLeptin Inversely correlated with GHNeuromedin C UnknownNeuropeptide Y ↓?Nitrc oxide No effectObesity ↓Opiates ↑Senescence/aging ↓Serotonin ↑Starvation ↑Stress (shock, restraint, endotoxin, psychological) ↑Testosterone ↑TRH No effectDHT No effect

DHT, dihydrotestosterone; GHRH, growth hormone releasing hormone; GHRP, growth hormone releasing peptide;GNRH, growth hormone releasing hormone; IGF-I, insulin-like growth factor I; SS, somatostatin; TRH, thyrotropinreleasing hormone.

Page 130: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

114 chapter 9

synthesis in skeletal muscle, resulting in hypertrophyof both muscle fiber types (Noall et al. 1957; Ullman& Oldfors 1989; Crist et al. 1991). Cartilage growthand bone deposition can also be stimulated by GH,increasing bone mineral density (BMD) and othermarkers of bone formation (Isaksson et al. 1990; vander Veen & Netelenbos 1990; Parfitt 1991; Bikle et al.1995; Orwoll & Klein 1995). While debatable withthe re-emergence of the larger family of GH(s) andGH binding proteins, classic dogma has proposedthat many of the anabolic effects of GH are mediatedvia IGF-I secreted by the liver and other tissues(Florini et al. 1996). While this may be true if GH isonly defined as the 22 kDa isoform, such a paradigmmay have to be re-examined if the GH superfamilyof polypeptides and binding proteins are now con-sidered. Nevertheless, exercise-induced GH releaseis in part responsible (either directly or indirectly)for the anabolic effects of exercise. Furthermore,there is evidence in rats that GH may stimulate theautocrine/paracrine production of IGF-I by skeletalmuscle, cartilage, and bone cells themselves (Turneret al. 1988; Isaksson et al. 1990; Bikle et al. 1995).Lastly, GH may interact both directly and indirectlywith androgens (Jørgensen et al. 1996), estrogens(Holmes & Shalet 1996) and thyroid hormone(Weiss & Refetoff 1996) with respect to secretion andtarget tissue actions.

Resistance exercise-induced changes ingrowth hormone concentrations

It has become apparent over the past 15 years thatdifferent exercise protocols will result in differentconcentrations of GH. To understand the compositefactors that may mediate such differential responseto the myriad of different exercise protocols possiblewith resistance training, one must look at some ofthe key factors that result from choices made in theacute program variables in workout design (Fleck &Kraemer 2004). It is also apparent that interaction ofvarious exercise factors is vital in determining themagnitude of the GH response. The key externalfactors that interact and produce an increased con-centration of GH in the circulatory biocompartmentare:1 The amount of muscle mass recruited.

2 The resistance loading used in the exercise.3 The volume of exercise performed.4 The amount of rest between sets and exercises.The activation of an adequate amount of muscle tis-sue is vital to increase plasma concentrations of GH.The amount of tissue that is activated is influencedby the resistance used, the total work and the type of exercise performed (e.g. small muscle group exercise vs. large muscle group exercise). The firstdata supporting this paradigm were observed byVanhelder et al. (1984). In their study, GH signific-antly increased above rest after performing sevensets with a resistance of 85% of the 7-repetition maximum (RM) in the squat exercise. However,when the resistance was reduced to 28% of the 7-RMwhile keeping rest periods the same and equatingthe total work, no changes in circulating concentra-tions of GH were observed after the exercise pro-tocol. Based on ‘size principle’, the number of motorunits required to perform the 28% load was less thanneeded for the 85% load. Thus, activation of enoughtissue appears to be a vital element of the exercisestimulus to elicit a significant GH response.

The volume of exercise or total work has also beenimplicated in the magnitude of response. Wheneach of the acute program variablesachoice of exercise, order of exercise, rest periods, resistanceusedaare kept constant and only when the numberof sets performed is increased thereby producingmore total work, is the response of GH increased.This effect has been demonstrated in both men andwomen using whole body, multiexercise resistanceprotocols and 10-RM loadings (Mulligan et al. 1996;Gotshalk et al. 1997). In addition, a higher level ofstrength fitness can also allow an individual to per-form greater amounts of total work resulting in ahigher GH response as well (Ahtiainen et al. 2003).Little is known about the thresholds of total work to elicit an increase in GH in the circulation, butlikely would be interactive with the other elementsof a workout protocol (e.g. length of rest periodsamount of tissue activated, and resistance used).

The interactive effects of different choices madefor the acute program variables (i.e. choice of exer-cise [e.g. large or small muscle groups], order ofexercise [e.g. large muscle group first or small mus-cle groups first], rest period length [short duration

Page 131: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 115

or long duration], resistance used [e.g. 5-RM or 10-RM] and number of sets or total work [low J orhigh J]) in designing a resistance exercise sessioncan result in a different GH response. While thecombinations that could be used are many, priorresearch has shown that when comparing multi-exercise, total body resistance protocols the threefactors that appear to impact GH most dramaticallyare the combination of high total work, short restperiod length (1 min between sets and exercise) anda moderately heavy 10-RM resistance in both menand women (Kraemer et al. 1990, 1993).

Since any resistance training session uses a widearray of different combinations of the acute exercisevariables, GH responses are a function of theseacute program variable choices. These appear to bethe underlying governing principles for the crea-tion of the exercise stimulus in resistance training.The most dramatic finding related to these variablestructures and GH is the impact they have on acid-base balance, which in turn appears to have amajor role in stimulating the release of GH into thecirculation. Each of the above four factors can bemanipulated to produce an effect on metabolismthat either limits or promotes the accumulation of hydrogen ions and decreases in blood pH, whichin turn account for almost half of the shared vari-ance with GH production. This makes acid-basedecreases (i.e. increase in ATP hydrolysis, decreasein pH, increase in hydrogen ion) prime determin-ants of the amount of 22 kDa isoform in the circula-

tion (Gordon et al. 1994). Reductions in the rest periods between sets of exercises with whole bodyworkouts have been shown to produce the mostdramatic increases in lactate responses with resist-ance exercise (Kraemer et al. 1990, 1993). However,reducing the rest period length will also impact theamount of resistance that can be lifted (Kraemer et al. 1987). Thus, there appears to be a crucial modu-lation of the amount of resistance used and thenamount of tissue activated that drives the GH res-ponse. However, Takarada et al. (2000) has shownthat occlusion of the arm can have a dramatic effecton GH resulting in significant increases with relat-ively low intensity (20% of 1-RM) while no changesin GH were observed without occlusion. One mayconclude that for the 22 kDa molecule, hypoxia anddisruption of acid-base balance plays a major regu-latory role in the stimulation of GH (Sutton 1983).Resistance exercise workouts that have short restperiods (1 min rest between sets and exercises, mod-erate intensities [8–10-RM load ranges] and wholebody workout protocols [8–10 exercises]) can pro-duce such physiological demands and results in themost dramatic GH changes in the blood (Fig. 9.3).

The majority of these studies have examined theshort-term recovery responses (typically < 2 h) ofGH. The impact of GH pulsatile release at differenttimes of day and GH roles over different phases ofrecovery from resistance exercise remains to be elucidated. McMurray et al. (1995) presented datawhich utilized a resistance exercise protocol as the

25

20

15

10

5

0

5-RM/3 min10-RM/1 min10-RM/3 min

Rest Mid 0 15 30 60

Fig. 9.3 GH responses to differentrest period lengths with differentresistance exercise protocols. The 10/1 post-exercise responses p < 0.05 from the other protocols ateach time point. All post-exerciseconcentrations are significantly (P < 0.05) above resting values.

Page 132: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

116 chapter 9

exercise perturbation on nocturnal GH responses.The exercise protocol consisted of performing 3 ×6–8-RM sets of six different exercises (18 sets total).Blood samples were obtained from 2100 to 0700 h.The investigators did not observe any effect of resistance exercise on nocturnal GH release. Con-versely, Nindl et al. (2001) examined GH pulsatileGH release over a longer period of time. The acuteheavy resistance exercise protocol began at 1500 hand was designed to be a high-volume workout thatincluded 50 total sets and recruited and activated alarge amount of muscle tissue. Blood was sampledevery 10 min from 1700 to 0600 h under control andresistance exercise conditions. The results of thestudy demonstrated that heavy resistance exercisein the late afternoon decreased overnight maximumGH concentrations and GH pulse amplitude; how-ever, overall mean GH concentrations were not sig-nificantly reduced. Acute heavy resistance exercisedifferentially influenced the temporal pattern of the overnight release because GH was lower duringthe first half of sleep, but greater during the secondhalf for the exercise versus control conditions. Themean maximum GH concentration and mean pulseamplitude were lower in the exercise vs. the controlcondition.

Several possible explanations for this findingwere speculated in that such results could be mediated by an increase in SS tone after exercise.Although SS inhibits GH release, it does not negat-ively affect GH biosynthesis. This may be an import-ant concept because it may explain a ‘rebound’ inGH secretion after SS priming/withdrawal. Thenocturnal peaks were lower for the exercise vs. control conditions during 2300–0300 h, but higherduring 0300–0600 h. Thus, even though the meanGH concentration was similar between the controland exercise conditions, the temporal pattern of GHrelease was clearly influenced by daytime exercise.From a mechanistic perspective, the acute heavyresistance exercise bout may have resulted in an elevated SS tone during 2300–0300 h. During thistime, GH release was inhibited to some degree andconcurrently; GH biosynthesis was not inhibited. At ~ 0300 h, this SS tone was withdrawn, and GHmolecules biosynthesized and stored during the timewhen GH release was inhibited were then available

and readily released. These data also demonstratedthat enhanced IGF-I inhibitory feedback on pitu-itary GH release was unlikely because serum IGF-Iconcentrations did not differ between the controland exercise conditions. It is well know that an arrayof other metabolic and hormonal signals, such aschanges in GHRH, hexapeptides, or Ghrelin release,could also have mediated the observed GH response.

Influence of age

It has been shown that the acute GH response issomewhat limited in older individuals (Craig et al.1989; Pyka et al. 1992; Kraemer et al. 1999). A majorfactor contributing to this limited GH responsemaybe the magnitude of exertion displayed and theinability to perform as much total work in an exer-cise protocol. It was reported in an investigation byPyka et al. (1992) that lower blood lactate concentra-tions in the elderly during resistance exercise sup-ported the theory that the amount of effort exertedin a resistance exercise session may impact theresulting GH response. With the negative influenceof aging on buffering capacities and toleration ofacidosis a combination of factors could help explainthe reductions in exercise-induced GH followingacute resistance exercise (Godfrey et al. 2003). Short-term training (10–12 weeks) does not appear to alterthis response (Craig et al. 1989; Kraemer et al. 1999).Ten-weeks periodized resistance training programshowed no significant changes in GH for resting or exercise-induced concentrations for younger or older subjects (Kraemer et al. 1999). The lactateresponse to the acute resistance exercise was lowerin the older than in the younger men with use of thesame relative resistance, while increases in lacticacid do not change the acid-base balance (Robergs et al. 2004), it acts as an indication of reduced meta-bolic demands and a lower disruption of acid-basestatus. This may partially explain the lower post-exercise GH values in older individuals. Reductionsobserved in resting and exercise-induced concentra-tions of the 22 kDa GH concentrations do not appearto be changed with short-term training (Fig. 9.4).

There is only a rudimentary understanding of the effects of exercise and aging on the physiolo-gical mechanisms underlying ‘somatopause’ (i.e. the

Page 133: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 117

decrease within the GH–IGF-I system). Because thisendocrine axis is considered to be of great import-ance in maintaining the integrity of the muscu-loskeletal system, it is conceptually pragmatic tosuggest that resistance exercise regimens shouldattempt to target it. Keeping in mind that GH alsoexhibits a great deal of molecular heterogeneity, andthe standard radioimmunoassay (RIA) focuses onthe 22-kDa variant, this is an especially importantpoint for future studies, when it is considered thatthe higher molecular weight species of GH couldpossess greater biological activity and that the lackof changes or reductions in the immunoreactive GHmay not present the complete picture of the adapta-tional responses of GH variants to resistance exer-cise training. In other words, measurement of justthe 22 kDa isoform may not tell the whole story ofthe adaptive response of pituitary secretions of GH.

Training adaptations

To date, resistance training does not appear to affectresting concentrations of the 22 kDa GH isoform.Surprisingly, no changes in the resting concentra-tions of GH have been observed in response to resist-ance training in men and women of various ages(Kraemer et al. 1999; McCall et al. 1999; Häkkinen et al. 2000; Marx et al. 2001). Even long-term training

in competitive lifters and body builders have shownthe same responses of the 22 kDa GH molecule andshow no changes in the hypopituitary axis for rest-ing concentrations (Häkkinen et al. 1988; Ahtiainenet al. 2003) when compared to untrained or lessertrained individuals. These findings are consistentwith dynamic feedback mechanisms and pulsatilityof the GH molecule and the many roles it may playin the homeostatic control of other metabolic andrepair processes. McCall et al. (1999) did observe acorrelations between resting GH and the magnitudeof type I and type II muscle fiber hypertrophy (r =0.62 to 0.74, respectively). These relationships couldbe indicative of a role for repeated acute resistanceexercise-induced GH elevations on cellular adapta-tions in trained muscle. Changes in receptor sensit-ivity, differences in feedback mechanisms, IGF-Ipotentiation, binding protein interactions, stimula-tion of other molecular weight variants in the pitu-itary somatotrophs as well as diurnal variationsmay all interact to mediate 22 kDa GH responses.

Gender effects

The exercise-induced responses of women com-pared to men appear to be similar in the absolutemagnitude of the value achieved but the relativechanges from rest are smaller (Kraemer et al. 1991).

Seru

m la

ctat

e (m

mo

l·L–1

)20

15

10

5

0Pre IP 5

Pretraining(a)15 30 Pre IP 5

Post-training15 30

* *

*#

*#

*

*

#*#

*#

**

*#

*

*#

**

*

Seru

m g

row

th h

orm

on

e (µ

g·L

–1)

15

10

5

0Pre IP 5

Pretraining(b)15 30 Pre IP 5

Post-training15 30

*

**

*

#

**

30 years 62 years 30 years 62 years

Fig. 9.4 Responses (means + SD) of lactate (a) and growth hormone (b) after acute heavy resistance exercise test (AHRET)before and after 10 weeks of periodized strength and power training for 30-year-old and 62-year-old men. *, Significantlydifferent (P ≤ 0.05) from corresponding pre-exercise value. #, Statistically significant difference (P ≤ 0.05) between 30-year-old and 62-year-old men. s, Significantly different (P ≤ 0.05) from corresponding pretraining value.

Page 134: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

118 chapter 9

This also impacts the acute exercise responses. Some protocols (e.g. 5-RM, 3 min rest periods or lowtotal work protocols) which may only marginallyelevate GH concentrations in the circulation in menresult in no significant exercise-induced elevationsin women due to the higher resting concentrations(Kraemer et al. 1991, 1993). How such lower magni-tudes of GH responses to acute exercise influencethe adaptations of target tissues remains unknown.

It has been postulated that women’s higher rest-ing concentrations potentially compensate for lowerlevels of other anabolic hormones which may min-imize the role of the acute elevation with exercisestress. These have been observed in the early follicu-lar phase of the menstrual cycle. Our recent dataindicate that estrogen in the form of oral contracept-ives may have minimal effects on the GH responseto resistance exercise response (unpublished data).While overt influences of the menstrual cycle maymediate some of these responses, differential pat-terns of pituitary GH release may also explain thegender differences as the GH mass and mode of GHsecretion may be regulated differently in men andwomen (Pincus et al. 1996).

Specificity of exercise

One of the underlying principles in biological sci-ence as well as a principle of resistance training is‘specificity’. This may be seen when examining theresponse of GH to resistance exercise as well. In astudy by Kraemer et al. (2001) four different groupsof subjects trained for 19 weeks performing all concentric repetitions, concentric repetitions withdouble the volume, or a typical program of con-centric and eccentric repetitions or no training (con-trol group). Subjects performed two exercise testsconsisting of three sets of 30 isokinetic concentriccontractions in one testing session and three sets of 30 isokinetic eccentric contractions with kneeextensions. The tests were separated by 48 h. Theacute response to a concentric stimulus was similar,but when an eccentric protocol was performed thegroup that had trained with the typical concentricand eccentric contraction style had the highest GHresponse to the eccentric challenge, indicating sensit-ivity to the specific eccentric stimulus that was con-

tained in the type of repetitions they were perform-ing with training. With detraining the responseswere muted to a similar degree across all groups.These data indicate that GH secretion may be sensit-ive to the specific muscle actions used during resist-ance training. Such a response is supported by therelatively new construct as it has been shown thatthe anterior pituitary may be directly innervated bynerve fibers mostly with synapses on corticotrophand somatotroph cells ( Ju 1999). It has also been suggested that ‘neural–humoral’ regulation of GHsecretion may take place such that a rapid neuralresponse is observed during the initial stress withthe humoral phase subsequently occurring (Ju 1999).If this is the case, then it may be possible for higherbrain centers (e.g. motor cortex) to play an activerole in regulating GH secretion during resistanceexercise and this regulatory mechanism may be sensitive to specific muscle actions used duringresistance training.

Physiological impact

Due to the many actions of GH and its complexity asa biological effector, its influence on skeletal musclehypertrophy and other tissue anabolism has juststarted to be dissected beyond simplistic direct andindirect hypotheses. A multitude of important inter-acting factors are thought to contribute to resistancetraining-induced skeletal muscle hypertrophy, andsome investigators have raised the possibility thatthe pronounced rise in blood GH concentrationelicited by acute heavy resistance exercise may beincluded with these mechanisms. This hypothesis issupported by observations that optimal heavy resist-ance training-induced skeletal muscle hypertrophyis compromised in hypophysectomized rats unlesssynthetic or pituitary-extracted GH is administered(Goldberg & Goodman 1969; Grindeland et al. 1994).However, directly contradicting this theory is thefact that an acute bout of aerobic exercise also pro-duces an equally large increase in blood GH concen-trations as an acute bout of heavy resistance exercisewhile aerobic exercise training has little or no netanabolic effect on skeletal muscle tissue (Kraemer et al. 1995). Therefore, assuming the GH response toexercise is important to muscle tissue hypertrophy

Page 135: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 119

and other tissue anabolism, it may be speculatedthat acute heavy resistance exercise and acute aerobic exercise differ not in their effect on serumimmunoreactive GH concentrations but in their effecton serum bioactive GH concentrations, an hypo-thesis that has recently received much attention(Hymer et al. 2001). Furthermore, the difference maybe due to activation of different motor neuron poolsand subsequent sequence of receptor events that arein part stimulated by the electromechanical activa-tion process of activated muscle fibers. Alternat-ively, immunoreactive GH may stimulate pituitaryrelease of higher molecular weight binding proteinsand aggregates into the circulation; each of thesehypotheses remains to be substantiated with directevidence (Nindl et al. 2003).

In addition, the pulsatile release of GH is import-ant for mediating linear growth. The growth processis episodic and discontinuous, whether measuredduring a period of a day or over longer periods oftime. For example, in rodents, linear growth is high-est when GH secretory bursts are separated byabout 3 h of very low GH levels, as in the case in themale rat, and is reduced when GH levels show smalldeviations around a relatively baseline, as in the caseof the female rat. Thus, sexually dimorphic mechan-isms regulating the pulsatile release of GH betweenmales and females (high amplitude GH pulses andlow interpulse [GH]) characterize GH profiles inmales, whereas female rats have less regular pulsesand higher interpulse [GH] are partly responsiblefor differences in growth rate (Slob & Van der WerffTen Bosch 1975; Jaffe et al. 1998). Additionally, thetemporal pattern of GH release is also coupled to key enzymes responsible for longitudinal bonegrowth and it may be important to view the GHrelease on a pulsatile basis rather than a single staticmeasure and this is well illustrated by consideringthe sexual dimorphism in GH release.

However, there are some animal data to suggestthat exercise-induced GH is important for somaticand muscle growth. Using the Hamster model,Katarina Borer has pioneered work into the role ofGH in exercise modulated growth by demon-strating that exercised golden hamsters (an animalcontinual growth model) exhibited increased basalpulsatile GH, skeletal elongation and permanentincrease in body mass and reductions in fat masswhen compared to sedentary controls (Borer 1989,1995). Since the pulsatile release of GH is an integralcomponent of agonist function, it would appear thatGH pulsatility in humans cannot be ignored as wellas the heterogeneity of the GH super family ofmolecules and aggregates. However, these werefemale hamsters and development takes place onlyat certain windows of a developmental time periodleading to maturation.

Summary

Acute resistance exercise can be a potent stimulusfor the 22 kDa GH isoforms with increases related to the type of exercise protocol utilized. Trainingappears to have no significant impact on restingconcentrations of these isoforms in men or women of different ages. However, changes in pulsatilerelease with acute exercise may be one mechanismby which an altered secretory profile may be medi-ated and such systems appear to differ between menand women. Feedback loops and interactions withother hormonal systems (e.g. IGF, GH binding pro-teins, stimulation of molecular aggregates) remaincomplex and an avenue for future research as itrelates to target tissue responses. It is evident thatthe responses and adaptations of GH will become a more integrated variable as the physiological context for its function and roles becomes furtherdelineated.

ReferencesAhtiainen, J.P., Pakarinen, A., Kraemer,

W.J. & Häkkinen, K. (2003) Musclehypertrophy, hormonal adaptations andstrength development during strengthtraining in strength-trained anduntrained men. European Journal ofApplied Physiology 89, 555–563.

Bikle, D.D., Harris, J., Halloran, B.P. et al.(1995) The molecular response of bone to growth hormone during skeletalunloading: regional differences.Endocrinology 136, 2099–2109.

Borer, K.T. (1989) Exercise-inducedfacilitation of pulsatile growth hormone

(GH) secretion and somatic growth. In:Hormones and Sport, (Laron, Z. & Rogol,A., eds.). Serono Symposia Publications,New York: 21–40.

Borer, K.T. The effects of exercise ongrowth. (1995) Sports Medicine (Auckland,NZ) 20(6), 375–397.

Page 136: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

120 chapter 9

Cappa, M., Grossi, A., Benedetti, S. et al.(1993) Effect of the enhancement of thecholinergic tone by pyridostigmine onthe exercise-induced growth hormonerelease in man. Journal of EndocrinologicalInvestigation 16, 421–424.

Craig, B.W., Brown, R. & Everhart, J. (1989)Effects of progressive resistance trainingon growth hormone and testosteronelevels in young and elderly subjects.Mechanisms of Ageing and Development49, 159–169.

Crist, D.M., Peake, G.T., Loftfield, R.B.,Kraner, J.C. & Egan, P.A. (1991)Supplemental growth hormone altersbody composition, muscle proteinmetabolism and serum lipids in fitadults: characterization of dose-dependent and response-recoveryeffects. Mechanisms of Ageing andDevelopment 58, 191–205.

Fleck, S.J. & Kraemer, W.J. (2004) DesigningResistance Training Programs, 3rd edn.Human Kinetics, Champaign, IL.

Florini, J.R., Ewton, D.Z. & Coolican, S.A.(1996) Growth hormone and the insulin-like growth factor system in myogenesis.Endocrine Reviews 17(5), 481–517.

Giustina, A. & Veldhuis, J.D. (1998)Pathophysiology of the neuroregulationof growth hormone secretion inexperimental animals and the human.Endocrine Reviews 19(6), 717–797.

Godfrey, R.J., Madgwick, Z. & Whyte, G.P.(2003) The exercise-induced growthhromeon response in athletes. SportsMedicine (Auckland, NZ) 33(8), 599–613.

Goldberg, A.L. & Goodman, H.M. (1969).Relationship between growth hormoneand muscular work in determiningmuscle size. Journal of Physiology 200,655–666.

Gordon, S.E., Kraemer, W.J., Vos, N.H.,Lynch, J.M. & Knuttgen, H.G. (1994)Effect of acid-base balance on the growthhormone resonse to acute high-intensitycycle exercise. Journal of AppliedPhysiology 76(2), 821–829.

Gotshalk, L.A., Loebel, C.C., Nindl, B.C. et al. (1997) Hormonal responses ofmultiset versus single-set heavy-resistance exercise protocols. CanadianJournal of Applied Physiology 22(3),244–255.

Grindeland, R.E., Roy, R.R., Edgerton, V.R.et al. (1994). Interactive effects of growthhormone and exercise on muscle mass in suspended rats. American Journal ofPhysiology 267, R316–R322.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi, P.V. (1988)Neuromuscular and hormonal

Kraemer, W.J., Fleck, S.J., Dziados, J.E. et al. (1993) Changes in hormonalconcentrations after different heavy-resistance exercise protocols in women.Journal of Applied Physiology 75(2),594–604.

Kraemer, W.J., Patton, J.F., Gordon, S.E. et al. (1995). Compatability of high-intensity strength and endurancetraining on hormonal and skeletalmuscle adaptations. Journal of AppliedPhysiology 78(3), 976–989.

Kraemer W.J., Fleck, S.J. & Evans, W.J.(1996) Strength and power training:physiological mechanisms ofadaptation. Exercise and Sport SciencesReviews 24, 363–397.

Kraemer, W.J., Häkkinen, K. Newton, R.U. et al. (1999) Effects of heavy-resistance training on hormonalresponse patterns in younger vs. oldermen. Journal of Applied Physiology 87,982–992.

Kraemer, W.J., Dudley, G.A., Tesch, P.A.et al. (2001) The influence of muscleaction on the acute growth hormoneresponse to resistance exercise andshort-term detraining. Growth Hormoneand IGF Research 11, 75–83.

McCall, G.E., Byrnes, W.C., Fleck, S.J. &Kraemer, W.J. (1999) Acute and chronichormonal responses to resistancetraining designed to promote musclehypertrophy. Canadian Journal of AppliedPhysiology 24, 96–107.

McMurray, R.G., Eubank, T.K. & Hackney, A.C. (1995) Nocturnalhormonal responses to resistanceexercise. European Journal of AppliedPhysiology 72, 121–126.

Marx, J.O., Ratamess, N.A., Nindl, B.C. et al.(2001) Low-volume circuit versus high-volume periodized resistance training in women. Medicine and Science in Sportsand Exercise 33(4), 635–643.

Mulligan, S.E., Fleck, S.J., Gordon, S.E. et al.(1996) Influence of resistance exercisevolume on serum growth hormone andcortisol concentrations in women.Journal of Strength and ConditioningResearch 10(4), 256–262.

Nindl, B.C., Hymer, W.C., Deaver, D.R. &Kraemer, W.J. (2001) Growth hormonepulsatility profile characteristicsfollowing acute heavy resistanceexercise. Journal of Applied Physiology91(1), 163–172.

Nindl, B.C., Kraemer, W.J., Marx, J.O.,Tuckow, A.P. & Hymer, W.C. (2003)Growth hormone molecularheterogeneity and exercise. Exercise andSport Sciences Reviews 31(4), 161–166.

adaptations in athletes to strengthtraining in 2 years. Journal of AppliedPhysiology 65, 2406–2412.

Häkkinen, K., Pakarinen, A., Kraemer,W.J., Newton, R.U. & Alen, M. (2000)Basal concentrations and acuteresponses of serum hormones andstrength development during heavyresistance training in middle-aged andelderly men and women. Journals ofGerontology. Series A, Biological Sciencesand Medical Sciences 55, B95–B105.

Holmes, S.J. & Shalet, S.M. (1996) Role ofgrowth hormone and sex steroids inachieving and maintaining normal bonemass. Hormone Research 45, 86–93.

Hunter, W.M., Fonseka, C.C. & Passmore,R. (1965) Growth hormone: importantrole in muscular exercise in adults.Science 150(699), 1051–1053.

Hymer, W.C., Kraemer, W.J., Nindl, B.C. et al. (2001) Characteristics of circulatinggrowth hormone in women after acuteheavy resistance exercise. AmericanJournal of Physiology. Endocrinology andMetabolism 281(4), E878–887.

Isaksson, O.G.P., Nilsson, A., Isgaard, J. &Lindahl, A. (1990) Cartilage as a targettissue for growth hormone and insulin-like growth factor I. Acta PaediatricaScandinavica. Supplement 367, 137–141.

Jaffe, C.A., Ocampo-Lim, B., Krueger, K. et al. (1998) Regulatory mechanisms of growth hormone secretion aresexually dimorphic. Journal of ClinicalInvestigation 102, 153–164.

Jørgensen, J.O.L., Vahl, N., Hansen, T.B. et al. (1996) Influence of growth hormone and androgens on bodycomposition in adults. Hormone Research45, 94–98.

Ju, G. (1999) Evidence for direct neuralregulation of the mammalian anteriorpituitary. Clinical ExperimentalPharmacology and Physiology. Supplement26, 757–759.

Kraemer, W.J., Noble, B.J., Clark, M.J. & Culver, B.W. (1987) Physiologicresponses to heavy-resistance exercisewith very short rest periods. InternationalJournal of Sports Medicine 8(4), 247–252.

Kraemer, W.J., Marchitelli, L., Gordon, S.E.et al. (1990) Hormonal and growth factorresponses to heavy resistance exerciseprotocols. Journal of Applied Physiology69(4), 1442–1450.

Kraemer, W.J., Gordon, S.E., Fleck, S.J. et al.(1991) Endogenous anabolic hormonaland growth factor responses to heavyresistance exercise in males and females.International Journal of Sports Medicine12(2), 228–235.

Page 137: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and resistance exercise 121

Noall, M.W., Riggs, T.R., Walker, L.M. &Christensen, H.N. (1957) Endocrinecontrol of amino acid transfer:distribution of an unmetabolizableamino acid. Science 126, 1002–1005.

Orwoll, E.S. & Klein, R.F. (1995)Osteoporosis in men. Endocrine Reviews16(1), 87–116.

Parfitt, A.M. (1991) Growth hormone andadult bone remodelling. ClinicalEndocrinology 35, 467–470.

Pincus, S.M., Gevers, E.F., Robinson, C. et al. (1996) Females secrete growthhormone with more process irregularitythan males in both humans and rats.American Journal of Physiology 270(1 Pt. 1),E107–E115.

Pyka, G, Wiswell, R.A. & Marcus, R. (1992)Age-dependent effect of resistanceexercise on growth hormone secretion inpeople. Journal of Clinical Endocrinologyand Metabolism 75, 404–407.

Robergs, R.A., Ghiasvand, F. & Parker, D.(2004) Biochemistry of exercise-inducedmetabolic acidosis. American Journal ofPhysiology. Regulatory, Integrative andComparative Physiology 287(3), R502–516.

Ullman, M. & Oldfors, A. (1989) Effects ofgrowth hormone on skeletal muscle. I.Studies on normal adult rats. ActaPhysiologica Scandinavica 135, 531–536.

Vanhelder, W.P., Radomski, M.W. &Goode, R.C. (1984) Growth hormoneresponses during intermittent weightlifting exercise in men. European Journalof Applied Physiology and OccupationalPhysiology 53(1), 31–34.

van der Veen, E.A. & Netelenbos, J.C.(1990) Growth hormone (replacement)therapy in adults: bone and calciummetabolism. Hormone Research 33 (suppl.4), 65–68.

Veldhuis, J.D., Patrie, J., Wideman, L. et al.(2004) Contrasting negative-feedbackcontrol of endogenously driven andexercise-stimulated pulsatile growthhormone secretion in women and men.Journal of Clinical Endocrinology andMetabolism 89(2), 840–846.

Weiss, R.E. & Refetoff, S. (1996) Effect ofthyroid hormone on growth: lessonsfrom the syndrome of resistance tothyroid hormone. Endocrinology andMetabolism Clinics of North America 25(3),719–730.

Scanlon, M.F., Isa, B.G. & Dieguez, C.(1996) Regulation of growth hormonesecretion. Hormone Research 46, 149–154.

Slob, A.K. & Van der Werff Ten Bosch, J.J.(1975) Sex differences in body growth inthe rat. Physiology and Behavior 14,353–361.

Snyder, G., Hymer, W.C. & Snyder, D.J.(1977) Functional heterogeneitysomatotrophs isolated from the ratanterior pituitary. Endocrinology 101(3),788–799.

Sutton, J.R. (1983) The hormonal responsesto exercise at sea level and at altitude.Progress in Clinical and Biological Research136, 325–341.

Takarada, Y., Nakamura, Y., Aruga, S. et al.(2000) Rapid increase in plasma growthhormone after low-intensity resistanceexercise with vascular occlusion. Journalof Applied Physiology 88(1), 61–65.

Turner, J.D., Rotwein, P., Novakofski, J. & Bechtel, P.J. (1988) Induction ofmRNA for IGF-I and -II during growthhormone-stimulated musclehypertrophy. American Journal ofPhysiology 255, E513–E517.

Page 138: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

122

Introduction

Growth hormone (GH) is secreted by the anteriorpituitary in a pulsatile pattern. Multiple GH iso-types and oligomers exist in plasma in addition tothe predominant 22 kDa protein (Baumann 1991;Lewis et al. 2000; Nindl et al. 2003). Minor isoformsdo not change uniquely in response to exercise. GH activates cells by dimerizing receptors and trig-gering a cascade of phosphorylation reactions thatsignal to the nucleus.

The amount of GH secreted in each pulse is underphysiological control by peptidyl agonists and ant-agonists (Arvat et al. 1998; Mueller et al. 1999; Farhyet al. 2001; Bowers 2002). Brain (hypothalamic)growth hormone releasing hormone (GHRH) stimu-lates GH synthesis and secretion and somatostatin(SS) inhibits GH release without affecting its syn-thesis (Giustina & Veldhuis 1998; Hartman 2000). Agrowth hormone releasing peptide (GHRP), ghre-lin, expressed in the stomach, anterior pituitarygland and hypothalamus amplifies GH secretion via cognate receptor codistributed with the pep-tide (Giustina & Veldhuis 1998; Kojima et al. 1999;Hartman 2000). Transgenic inactivation of cent-ral–neural GHRP receptors reduces GH secretion by one-third in the mouse (Shuto et al. 2002). Thesethree effector molecules govern GH secretion byconvergent mechanisms (Veldhuis & Bowers 2003b).Many of the metabolic effects of GH are mediatedby insulin-like growth factor I (IGF-I), which is synthesized in the liver and all nucleated cells underthe control of GH and tissue-specific hormones(Giustina & Veldhuis 1998).

GH secretion declines by approximately 14% perdecade after the age of 40 years (Rudman et al. 1981;Zadik et al. 1985; Iranmanesh et al. 1991) and ismarkedly reduced in obesity, even in younger indi-viduals (Veldhuis et al. 1991, 1995). Whereas GHproduction falls by 50% every 7 years in men begin-ning in young adulthood (Iranmanesh et al. 1991,1998; Veldhuis et al. 1995), the decrease is nearlytwofold less rapid in premenopausal women(Asplin et al. 1989; Winer et al. 1990; Weltman, A. etal. 1994; van den Berg et al. 1996). Many age-relatedphysical adaptations resemble those recognized inGH-deficient adults, including reduced musclemass and exercise capacity, increased body fat espe-cially abdominal visceral fat, unfavorable lipid andlipoprotein profiles, reduction in bone mineral dens-ity and cerebro and cardiovascular disease. Which is cause and which is effect is difficult to ascertain, inthat intra-abdominal adiposity and limited exercisealso predict reduced GH production (Vahl et al.1997; Clasey et al. 2001). Below we highlight theeffects of exercise on GH release. Several compre-hensive reviews discuss other physiological factors(Veldhuis 1996a, 1996b; Veldhuis et al. 1997; Giustina& Veldhuis 1998; Hartman 2000; Veldhuis & Bowers2003a, 2003b).

Acute aerobic exercise and growthhormone secretion

Aerobic exercise stimulates GH release withinapproximately 15 min and induces peak values at ornear the end of exertion (Lassarre et al. 1974; Sutton& Lazarus 1976; Kozlowski et al. 1983; Raynaud et al.

Chapter 10

The Growth Hormone Response to Acute andChronic Aerobic Exercise

ARTHUR L. WELTMAN, LAURIE WIDEMAN, JUDY Y. WELTMAN AND

Page 139: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 123

1983; Bunt et al. 1986; Chang et al. 1986; Felsing et al.1992; Luger et al. 1992; Weltman, A. et al. 1992, 1994;Cappon et al. 1994; Chwalbinska-Moneta et al. 1996;Pritzlaff et al. 1999, 2000; Wideman et al. 1999,2000a). The intensity and duration of the aerobicstress, physical fitness, gender and age all influencethe GH response to exercise. Although the threshold of exercise intensity was envisioned earlier (Changet al. 1986; Felsing et al. 1992; Chwalbinska-Monetaet al. 1996), randomly ordered separate day studiesaffirm that exercise intensity predicts GH secretionin a linear dose–response fashion (Pritzlaff et al.2000; Pritzlaff-Roy et al. 2002).

Women maintain higher GH concentrations thanmen at all ages, and manifest less orderly patterns ofpulsatile GH release (Hartman et al. 1990; Veldhuis1995; van den Berg et al. 1996; Pincus et al. 1996;Engstrom et al. 1998; Giustina & Veldhuis 1998;Wideman et al. 1999). The time course of exercise-induced GH release is similar by gender (Lassarre et al. 1974; Bunt et al. 1986), but women differ in sev-eral specific features; viz: anticipatory GH releasebefore exercise and more rapid attainment of peakGH concentrations during exercise (Wideman et al.1999, 2000a; Pritzlaff-Roy et al. 2002). The effect of exercise is largely independent of circadianrhythmicity, since the time of day does not influencethe responses, at least in young men (Kanaley et al.2001). Young women and men achieve equival-ent absolute GH concentrations during exercise(Wideman et al. 1999), but the fractional increaseover baseline is higher in men (Bunt et al. 1986;Wideman et al. 2000a).

Figure 10.1 illustrates the impact of gender onexercise intensity-dependent GH release in youngadults (Pritzlaff et al. 1999; Pritzlaff-Roy et al. 2002).In this study, men and women each undertook sixrandomly ordered sessions (one control resting [C]and five exercise conditions [Ex]). Exercise com-prised 30 min of treadmill running at one of the following graded intensities (normalized to theindividual lactate threshold [LT]): 25% and 75% ofthe difference between LT and rest (0.25 LT and 0.75LT, respectively), LT, and 25% and 75% of the differ-ence between LT and Vo2peak (1.25 LT and 1.75 LT,respectively). GH responses to exercise increasedprogressively with increasing exercise intensity

(Pritzlaff et al. 1999; Pritzlaff-Roy et al. 2002). By simple linear regression analysis, women had ahigher intercept value (greater baseline GH secre-tion) and slope (accentuated sensitivity) to aerobicexercise (Fig. 10.2).

The extent that these acute relationships apply tograded chronic exercise training intensities is notknown.

Exercise stimulates less GH secretion in healthyolder individuals than in young individuals(Weltman, A. et al. 2000b, 2001). Analogous exerciseintensity–GH response comparisons indicate thataging in man attenuates the graded effectiveness of exercise (slope term by 3.9-fold). An unresolvedquestion raised by these outcomes is whether higherrelative training intensities would be able to driveyoung adult-like GH release in older men (Weltman,A. et al. 2000b).

Exercise stimulates GH secretion 5.7–7.3-fold lessin post-menopausal women than in premenopausalwomen, whether or not post-menopausal individu-als were receiving hormone replacement (Marcell et al. 1999; Weltman, A. et al. 2001). Plausible mech-anistic bases for the consistent deficit in GH secre-tion in the older adult include excessive SS releaseand diminished GHRH or possibly ghrelin drive. In fact, prolonged stimulation with GHRH or a synthetic GHRP (ghrelin surrogate) will elevate GHsecretion over 1–3 months in the elderly individual(Evan et al. 2001; Richmond et al. 2001).

The GH response to exercise may also be bluntedin middle-aged men (Zaccaria et al. 1999). This infer-ence derives from study of a small group of young(N = 5, age = 21 years) and middle-aged (N = 7, age = 42 years) men who undertook incrementalexercise (50 watts every 3 min) until volitionalexhaustion (Fig. 10.3).

Middle-aged men evidence lower and delayedpeak GH release. If verified, diminutive GH responsiveness before later life would encourageinvestigations of earlier interventional strategies tomaintain favorable GH drive of anabolism.

Relative adiposity and frank obesity suppressbaseline and stimulated GH production (Veldhuiset al. 1995; Weltman, A. et al. 2000b; Weltman, J.Y. et al. 2002). When compared to various pharmaco-logic and physiologic stimuli, the magnitude of the

Page 140: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

124 chapter 10

1600

1200

800

400

017515012510075

Exercise intensity (% LT)

50250

Exer

cise

& r

eco

very

(09

00–1

300

h)

Inte

gra

ted

ser

um

GH

(µg

·L−1

·min

−1)

Females (P = 0.02)X Slope = 449X Intercept = 562

Males (P = 0.08)X Slope = 277X Intercept = 198

Fig. 10.2 Impact of gender onbaseline (unstimulated) and exercise-induced growth hormone (GH)secretion assessed over a gradedrange of intensity (see Fig. 10.1). Thehigher slope of this relationship inwomen denotes a greater sensitivityof the GH response to exercise, andthe elevated intercept signifies higherbaseline (rest) GH secretion. (FromPritzlaff-Roy 2002, used withpermission.)

Mea

n s

eru

m G

H (

µg. L

–1)

30

25

20

15

10

5

0

Males(N = 10)

Onset ofexercise

Control0.25 LT0.75 LTLT1.25 LT1.75 LT

13:0012:2011:4011:0010:209:409:008:207:407:00

30

25

20

15

10

5

0

Females(N = 8)

Onset ofexercise

Control0.25 LT0.75 LTLT1.25 LT1.75 LT

13:0012:2011:4011:0010:20Time

9:409:008:207:407:00

Mea

n s

eru

m G

H (

µg. L

–1)

Fig. 10.1 Mean serum growthhormone (GH) concentrationsmonitored by sampling blood every10-min for 6 h during rest (Control),and graded exercise defined asfractions (0.25, 0.75, 1.0, 1.25 and 1.75)of the individual lactate threshold(LT). Values are the mean ± SEM (N = 10 young men and eight youngwomen). (From Pritzlaff et al. 1999and Pritzlaff-Roy 2002, used withpermission.)

Page 141: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 125

GH response to exercise in obese subjects may exceedthat induced by l-dopa or clonidine (Cordido et al.1990; Tanaka et al. 1990), but not necessarily GHRH,pyridostigmine or l-arginine (Williams et al. 1984;Cordido et al. 1990, 1993; Maccario et al. 1997;Kelijman & Frohman 1998). Only GHRP and com-bined secretagogues remain moderately (but notmaximally) effective (Cordido et al. 1990, 1993;Maccario et al. 1997). Limited analyses of the inter-action among gender, obesity and exercise suggestthat abdominal visceral fat may be a key determin-ant of stimulated GH release (Clasey et al. 2001).Kanaley et al. (1999) examined exercise induced GH secretion in cohorts comprising of non-obese,lower-body obese and upper-body obese (e.g. vis-cerally obese) women (Fig. 10.4).

1800

1200900

600

300

Seconds

60 180E0

80

70

60

50

40

30

20

10

0

GH

(µg

·L−1

)

Fig. 10.3 Growth hormone (GH)responses to maximal exercise atexhaustion (E) and during recoveryin young (diamonds) and middle-aged (circles) athletes before (open)and after (filled) a 4-month period oftraining (Adapted from Zaccaria et al.1999.)

15

5

0

(µg

·L−1

)

5310(a) –1

15

5

0

(µg

·L−1

)

5310(b)

(c)

–1

15

5

0

(µg

·L−1

)

300180600–1

Time (minutes)

Fig. 10.4 (right) Mean serum growth hormone (GH)concentrations for non-obese (a), lower body obese (b),and upper body obese (c) groups over the 6-h period of blood sampling. Exercise began at time zero andcontinued for 30 min at an intensity of 70% Vo2peak.(Adapted from Kanaley et al. 1999.)

Page 142: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

126 chapter 10

The rank order of peak GH concentrations was13.7 µg·L–1 (non-obese), 6.8 µg·L–1 (lower bodyobese) and 3.5 µg·L–1 (upper body obese). Definit-ive radiological measurement of adipose-tissuetopography will be needed to verify these anthro-pometric inferences.

Chronic endurance training and growthhormone release

Sustained endurance training limits acute exercise-induced GH release at the same absolute intensity(Hartley et al. 1972; Weltman, A. et al. 1997). In

Pre(a) Week 3

*

Post

*

400

300

200

100

0

Inte

gra

ted

[G

H]

(µg

·L−1

× m

in)

Pre(c) Week 3

*

Post

*

30

20

10

0

[No

rep

inep

hri

ne]

(nm

ol·L

−1)

Pre(b) Week 3 Post

* *

4000

3000

2000

1000

0

[Ep

inep

hri

ne]

(pm

ol·L

−1)

Fig. 10.5 The effects of 6 weeks of training on integratedgrowth hormone (GH) concentration (a) and end exerciseconcentrations of epinephrine (b) and norepinephrine (c)in response to a constant load cycle ergometry exercisebout (n = 6, Mean ± SD), Pre, Pretraining; Week 3, After 3 weeks of training; Post, After 6 weeks of training. *P < 0.05 vs. pretraining. (From Weltman et al. 1997, usedwith permission.)

Baseline

Time

******

1 year

C@ LT> LT

10 000

8000

6000

4000

2000

Inte

gra

ted

(24

h)

GH

con

cen

trat

ion

(µg

·L−1

·min

−1)

Fig. 10.6 Effects of 1-year of runtraining on 24-h integrated serum growth hormone (GH)concentrations. *1 year > baseline in > lactate threshold (LT) group (P < 0.05); **at 1 year > LT group >control (C) group (P < 0.05); ***at 1 year > LT group > @LT group (P < 0.05). (From Weltman et al. 1992,used with permission.)

Page 143: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 127

young men, response down-regulation is evidentwithin the first 3 weeks of training (Fig. 10.5)(Weltman, A. et al. 1997).

On the other hand, total (24-h) GH secretionappears to increase in trained individuals even onnon-training days when some aerobic training wastargeted at an intensity above the lactate threshold(Fig. 10.6) (Weltman, A. et al. 1992).

How healthy aging, gender and increased adi-

posity modulate long-term training effects is not yet established. In one study in older men (ages50–78 years), serum IGF-I concentrations did notdiffer between marathon runners and age-matchedsedentary subjects (Deuschle et al. 1998). In anotherinvestigation, 1 year of exercise training (eithersupervised aerobic training [4 days a week] orsupervised strength training [3 days a week]) inhealthy older (aged 59–79 years) adults did not alter24-h integrated GH concentrations (Hartman et al.2000). Apparent unresponsiveness might be due to: (i) an insufficient training stimulus; (ii) lack ofchange in percentage body fat and abdominal vis-ceral fat which correlate negatively with GH releaseand increase with age; and/or (iii) intrinsicallydiminished responsiveness of the aging GH–IGF-Iaxis.

Sixteen weeks of aerobic training in obese womenyielded a significant training effect (e.g. increasedVo2peak), but did not alter GH responses to acuteexercise at the same relative intensity (Fig. 10.7)(Kanaley et al. 1999). However, training for 4 monthsdid not affect body weight, fat weight and fat freeweight (estimated by skinfolds and bioelectricalimpedance). Whether daily GH secretion rates in-crease in this context is not known. Indeed, the pre-cise relationship between acute exercise-stimulated

600

300

0Inte

gra

ted

GH

co

nce

ntr

atio

ns

(6h

) (m

in·µ

g−1

·L−1

)

Pre-exercise

Post-exercise

Fig. 10.7 The 6-h integrated serum growth hormone (GH)concentrations in 10 obese subjects before and after 16weeks of aerobic exercise in response to an exercise bout at 70% Vo2peak for 30 min. (Adapted from Kanaley et al.1999.)

α2-Adrenergic

Exercise

Human GH axis: neuromodulators

Arginine

?

?

?

?

?

Dopamine

Cholinergic

β2-Adrenergic

Putative GHRPs

GHRH

GH

**

IGF-I

Somatostatin

Fig. 10.8 Schematic representation of the possible interactions ormechanisms that control exercise-induced growth hormone (GH)release. *, indicates the possibilitythat exercise modifies the normalautonegative feedback control of GH on growth hormone releasinghormone (GHRH) and somatostatin;+, denotes stimulation; –, denotesinhibition. (From Giustina &Veldhuis 1998. © 1998, The EndocrineSociety.)

Page 144: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

128 chapter 10

and training-enhanced (mean) GH production onnon-training days remains unclear.

Neuroendocrine control of exercise-induced growth hormone release

The neuroendocrine basis underlying exercise-induced GH release is complex and remains enigm-atic (Giustina & Veldhuis 1998; Wideman et al. 2000a).Presumptive mechanisms involve GHRH, SS and/or ghrelin (Fig. 10.8) (Veldhuis & Bowers 2003b).

Putative modulators include (non-exclusively)catecholamines, muscarinic agonists, opiatergic pep-

tides, GABA and possibly excitatory amino acids(Thompson et al. 1993; Giustina & Veldhuis 1998;Weltman, A. et al. 2000a). Albeit correlational, plasmanorepinephrine concentrations peak before and areproportionate to exercise-stimulated GH concentra-tions in young men with and without exercise training (Weltman, A. et al. 1997, 2000a). Such dataare consistent with, but not proof of, central nora-drenergic (α2-adrenorecptor) drive. Serum ghrelinconcentrations are higher in lean than obese adults(Veldhuis & Bowers 2003b), but do not change during 45 min of exercise and 3 h of recovery (Dall et al. 2002).

160

140

120

100

80

60

40

20

0

Seru

m G

H c

on

cen

trat

ion

(µg

·L−1

)

700600 900

Clock time(a)

1000 1100 1200800

SAGAG

160

140

120

100

80

60

40

20

0

Seru

m G

H c

on

cen

trat

ion

(µg

·L−1

)

700600 900

Clock time(b)

1000 1100 1200800

SAGAG

Fig. 10.9 Mean serum growthhormone (GH) concentration (µg·L–1)profiles basally and in response tosaline (S), GH releasing peptide-2 (G),and/or l-arginine (A) infusion at restin men (a) and women (b). Data aremeans ± SEM. Clock time (hours) isshown. (From Wideman et al. 2000b,used with permission.)

Page 145: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 129

Two probes of neuroendocrine pathways mediat-ing GH secretion are l-arginine and GHRP-2, whichinduce SS withdrawal and mimic ghrelin actionrespectively. GHRP-2 also potentiates the effect ofGHRH and mutes that of SS (Bowers et al. 1994;Pihoker et al. 1995; Popovic et al. 1995; Giustina & Veldhuis 1998; Bowers & Granda-Ayala 1999;Mueller et al. 1999; Veldhuis & Bowers 2003b). Allthree agonists show sensitivity to gender (Merimeeet al. 1969; Benito et al. 1991; Bercu et al. 1991; Bowers1993; Penelva et al. 1993; Veldhuis 1996a, 1998, 2003;Giustina & Veldhuis 1998; Jaffe et al. 1998; Veldhuiset al. 2001; Veldhuis & Bowers 2003a). At rest, basal

and l-arginine (but not GHRP-2) stimulated GHrelease is higher in women than men (Fig. 10.9)(Wideman et al. 2000b).

The synergy is equivalent in absolute terms in the female and the male before exertion. Exercisepotentiates maximal GH concentrations driven byl-arginine or GHRP-2 alone as well as together, and absolute responses are compatible by gender(Fig. 10.10) (Wideman et al. 2000a).

Fractional responses to exercise (fold-increaseabove rest) are twofold higher in men than womenadministered combined stimuli. Since GHRH ex-pressly facilitates GHRP-2 and l-arginine actions,

120

100

80

60

40

20

0Se

rum

GH

co

nce

ntr

atio

n (

µg·L

−1)

700600 900

Clock time(a)

1000 1100 1200800

SAGAG

120

100

80

60

40

20

0

Seru

m G

H c

on

cen

trat

ion

(µg

·L−1

)

700600 900

Clock time(b)

1000 1100 1200800

SAGAG

Fig. 10.10 Mean serum growthhormone (GH) concentration (µg·L–1)profiles basally and in response tosaline (S), GH releasing peptide-2 (G),and/or l-arginine (A) infusion withexercise in men (a) and women (b).Data are means ± SEM. Clock time(hours) is shown. (From Wideman et al. 2000a, used with permission.)

Page 146: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

130 chapter 10

References

Arvat, E., Ceda, G.P., Di Vito, L. et al. (1998) Age-related variations in theneuroendocrine control, more thanimpaired receptor sensitivity, cause thereduction in the GH-releasing activity of GHRPs in human aging. Pituitary 1,51–58.

Asplin, C.M., Faria, A.C., Carlsen, E.C. et al. (1989) Alterations in the pulsatilemode of growth hormone release in menand women with insulin-dependentdiabetes mellitus. Journal of ClinicalEndocrinology and Metabolism 69,239–245.

Baumann, G. Growth hromoneheterogeneity: genes, isohormones,variants and binding proteins. EndocrineReviews 12, 424–449.

Benito, P., Avila, L., Corpas, M.S. et al.(1991) Sex differences in growthhormone response to growth hormone-releasing hormone. Journal ofEndocrinological Investigation 14, 265–268.

Bercu, B.B., Weideman, C.A. & Walker,R.F. (1991) Sex differences in growthhormone (GH) secretion by ratsadministered GH-releasinghexapeptide. Endocrinology 129,2592–2598.

van den Berg, G., Veldhuis, J.D., Frolich,M. et al. (1996) An amplitude-specificdivergence in the pulsatile mode of GHsecretion underlies the gender differencein mean GH concentrations in men andpremenopausal women. Journal ofClinical Endocrinology and Metabolism 81,2460–2466.

Bowers, C.Y. (1993) GH releasingpeptidesastructure and kinetics. Journalof Pediatric Endocrinology 6, 21–31.

Bowers, C.Y. (2002) New insight into thecontrol of growth hormone secretion. In: Central and peripheral Mechanisms inPituitary Disease (Kleinberg, D.L. &Clemmons, D.R., eds.). BioScientifica,Bristol, UK: 163–176.

Bowers, C.Y. & Granda-Ayala, R. (1999)Stimulated release of GH in normalyounger and older men and women. In: Sex-Steroid Interactions with GrowthHormone (Veldhuis, J.D. & Giustina, A.,eds.). Serono Symposia, Norwell, MA:277–289.

Bowers, C.Y., Maumenee, F.A., Reynolds,G.A. & Hong, A. (1984) On the in vitroand in vivo activity of a new synthetichexapeptide that acts on the pituitary to specifically release growth hormone.Endocrinology 114, 1537–1545.

Bowers, C.Y., Veeraragavan, K. &Sethumadhavan, K. (1994) Atypicalgrowth hormone releasing peptides. In: Growth Hormone. II. Basic ClinicalAspects (Bercu, B.B. & Walker, R.F., eds.). Springer-Verlag, New York:203–222.

Brill, K.T., Weltman, J.Y., Anderson, S. et al. (2002) Relative rank order ofdiscrete secretagogue actions in the

synergy of the latter two secretagogues during exer-cise strongly supports the inference that exertionreleases hypothalamic GHRH. This concept will needto be tested by selective GHRH-receptor antagonists.

In complementation, estimates of stimulated GHsecretory-burst mass (Veldhuis et al. 1990) corrobor-ate the impact of l-arginine and GHRP-2 (Widemanet al. 2000a, 2000b). However, GHRH, SS and put-atively endogenous ghrelin are comodulated byinhibitory (β-adrenergic and α1-noradrenergic) andstimulatory (cholinergic and opiatergic) signal (Ghigoet al. 1993, 1994; Thompson et al. 1993; Giustina &Veldhuis 1998; Mueller et al. 1999; Veldhuis &Yoshida 2000). Exercise appears to stimulate multiplecentral–neural neurotransmitters (Sutton & Lazarus1974; Uusitupa et al. 1982; Moretti et al. 1983; Bowerset al. 1984; Thompson et al. 1993; Giustina &Veldhuis 1998). The integration of such responses isa daunting challenge in exercise physiology.

An important emerging issue is the degree towhich specific GH secretagogues can amplify theeffects of exercise in aging individuals. In one pre-liminary study in older men, combined stimulationwith GHRP-2 and aerobic exercise elicited greaterGH secretion (summed amplitude) than eitherintervention alone, but did not act with true synergy

(supra-additive effect) (Weltman, A. et al. 2002). Thelater outcome could denote excessive somatosta-tinergic restraint, impaired outflow of endogenousGHRH (which synergizes with GHRP), and/ordown-regulation of the GHRP signaling pathway as inferred independently in the aging human andexperimental animal (Veldhuis et al. 2001, 2002;Veldhuis 2003). A corollary question posed by cur-rent data is how all three of exercise, age and gendergovern GH secretion (Brill et al. 2002).

Conclusions

Aerobic exercise is a potent stimulus for GH release,particularly in young men and women. Age andobesity blunt responsiveness markedly, putativelyby modifying metabolic signals to and effects of all three of GHR (stimulatory), SS (inhibitory) andghrlein (GHRP, amplifying). Gender further deter-mines GH secretion driven by exercise at any age. In principle, impoverishment of physiological GHsecretion in sedentary, obese and aged individualswould exacerbate visceral fat accumulation, dyslipi-demia, relative insulin resistance, diminished boneand muscle mass and (possibly) reduced quality oflife.

Page 147: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 131

healthy aged male. Paper presented at the 84th Annual Meeting of theEndocrine Society, San Francisco, CA.

Bunt, J.C., Boileau, R.A., Bahr, J.M.,Nelson, R.A. (1986) Sex and trainingdifferences in human growth hormoneduring prolonged exercise. Journal ofApplied Physiology 61, 1796–1801.

Cappon, J.P., Brasel, J., Mohan, S. et al.(1994) Effect of brief exercise oncirculation insulin-like growth factor I.Journal of Applied Physiology 76,2490–2496.

Chang, F.E., Dodds, W.G., Sullivan Kim,M.H. & Malarkey, W.B. (1986) The acute effects of exercise on prolactin andgrowth hormone secretion: comparisonbetween sedentary women and womenrunners with normal and abnormalmenstrual cycles. Journal of ClinicalEndocrinology and Metabolism 62,551–556.

Chwalbinska-Moneta, J., Krysztofiak, H.,Ziemba, A. et al. (1996) Thresholdincreases in plasma growth hormone in relation to plasma catecholamine andblood lactate concentrations duringprogressive exercise in endurance-trained athletes. European Journal ofApplied Physiology 73, 117–120.

Clasey, J.L., Weltman, A., Patrie, J. et al.(2001) Abdominal visceral fat andfasting insulin are important predictorsof 24-hour GH release independent ofage, gender and other physiologicalfactors. Journal of Clinical Endocrinologyand Metabolism 86, 3845–3852.

Cordido, F., Dieguez, C. & Casanueva, F.F. (1990) Effect of central cholinergicneurotransmission enhancement bypyridostigmine on the growth hormonesecretion elicited by clonidine, arginine,or hypoglycemia in normal and obesesubjects. Journal of Clinical Endocrinologyand Metabolism 70, 1361–1370.

Cordido, F., Penalva, A., Dieguez, C. et al.(1993) Massive growth hormone (GH)discharge in obese subjects after thecombined administration of GH-releasing hormone and GHRP-6:evidence for a marked somatotrophsecretory capability in obesity. Journal ofClinical Endocrinology and Metabolism 76,819–823.

Dall, R., Kanaley, J., Hansen, T.K. et al.(2002) Plasma ghrelin levels duringexercise in healthy subjects and ingrowth hormone-deficient patients.European Journal of Endocrinology 147,65–70.

Deuschle, M., Blum, W.F., Frystyk, J. et al.(1998) Endurance training and its effect

does not increase 24-h GH secretion inolder adults. Paper presented at the82nd Annual Meeting of the EndocrineSociety, Toronto, Canada.

Iranmanesh, A., Lizarralde, G. & Veldhuis, J.D. (1991) Age and relativeadiposity are specific negativedeterminants of the frequency andamplitude of growth hormone (GH)secretory bursts and the half-life ofendogenous GH in healthy men. Journalof Clinical Endocrinology and Metabolism73, 1081–1088.

Iranmanesh, A., South, S., Liem, A.Y. et al. (1998) Unequal impact of age,percentage body fat, and serumtestosterone concentrations on thesomatotrophic, IGF-I, and IGF-bindingprotein responses to a 3-day intravenousgrowth hormone-releasing hormonepulsatile infusion in men. EuropeanJournal of Endocrinology 139, 59–71.

Jaffe, C.A., Ocampo-Lim, B., Guo, W. et al. (1998) Regulatory mechanisms ofgrowth hormone secretion are sexuallydimorphic. Journal of ClinicalInvestigation 102, 153–164.

Kanaley, J.A., Weatherup-Dentes, M.M.,Jaynes, E.B. et al. (1999) Obesityattenuates the growth hormoneresponse to exercise. Journal of ClinicalEndocrinology and Metabolism 84,3156–3161.

Kanaley, J.A., Weltman, J.Y., Pieper, K.S. et al. (2001) Cortisol and growthhormone responses to exercise atdifferent times of day. Journal of ClinicalEndocrinology and Metabolism 86,2881–2889.

Kelijman, M. & Frohman, L.A. (1988)Enhanced growth hormone (GH)responsiveness to GH-releasinghormone after dietary manipulation inobese and nonobese subjects. Journal ofClinical Endocrinology and Metabolism 66,489–494.

Kojima, M., Hiroshi, H., Date, Y. et al.(1999) Ghrelin is a growth-hormonereleasing acylated peptide fromstomach. Nature 402, 656–666.

Kozlowski, S., Chwalbinska-Moneta, J.,Vigas, M. et al. (1983) Greater serum GHresponse to arm than to leg exerciseperformed at equivalent oxygen uptake.European Journal of Applied Physiology 52,11–135.

Lassarre, C., Girard, F., Durand, J. et al.(1974) Kinetics of human growthhormone during submaximal exercise.Journal of Applied Physiology 37, 826–830.

Lewis, U.J., Sinha, Y.N., Lewis, G.P. (2000)Structure and properties of members of

upon the activity of the GH-IGFs systemin the elderly. International Journal ofSports Medicine 19, 250–254.

Engstrom, B.E., Karlsson, F.A. & Wide, L.(1998) Marked gender differences inambulatory morning growth hormonevalues in young adults. ClinicalChemistry 44(6), 1289–1295.

Evan, W.S., Bowers, C.Y. & Veldhuis, J.D.(2001) Estradiol supplementationenhances pituitary sensitivity torecombinant human (RH) GHRH-1, 44-amide in somatostatin (SS)-withdrawn postmenopausal women.Paper presented at the EndocrineSociety 83rd Meeting, Denver, CO.

Farhy, L.S., Straume, M., Johnson, M.L. et al. (2001) A construct of interactivefeedback control of the GH axis in themale. American Journal of Physiology 281,R38–R51.

Felsing, N.E., Brasel, J.A. & Cooper, D.M.(1992) Effect of low and high intensityexercise on circulating growth hormonein men. Journal of Clinical Endocrinologyand Metabolism 75, 157–162.

Ghigo, E., Arvat, E., Bellone, J., Ramunni, J.& Camanni, F. (1993) Neurotransmittercontrol of growth hormone secretion inhumans. Journal of PediatricEndocrinology 6, 263–266.

Ghigo, E., Arvat, E., Gianotti, L. et al. (1994) Interaction of salbutamol withpyridostigmine and arginine on bothbasal and GHRH-stimulated GHsecretion in humans. ClinicalEndocrinology 40, 799–802.

Giustina, A. & Veldhuis, J.D. (1998)Pathophysiology of the neuroregulationof growth hormone secretion inexperimental animals and the human.Endocrine Reviews 19(6), 717–797.

Hartley, L.H., Mason, J.W., Hogan, R.P. et al. (1972) Multiple hormonalresponses to graded exercise in relationto physical training. Journal of AppliedPhysiology 33, 602–606.

Hartman, M.L. (2000) Physiologicalregulators of growth hormone secretion.In: Growth Hormone in Adults, 2nd edn.(Juul, A. & Jorgensen, J.O.L., eds.).Cambridge University Press,Cambridge, UK: 3–53.

Hartman, M.L., Veldhuis, J.D., Vance, M.L. et al. (1990) Somatotropin pulsefrequency and basal concentrations are increased in acromegaly and arereduced by successful therapy. Journal ofClinical Endocrinology and Metabolism 70,1375–1384.

Hartman, M.L., Weltman, J.Y., Patrie, J.T. et al. (2000) Exercise training for 1 year

Page 148: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

132 chapter 10

the hGH family: a review. EndocrineJournal 47 (suppl.), S1–S8.

Luger, A., Watschinger, B., Duester, P. et al. (1992) Plasma growth hormone andprolactin responses to graded levels ofacute exercise and to a lactate infusion.Neuroendocrinology 56, 112–117.

Maccario, M., Valetto, M.R., Savio, P. et al.(1997) Maximal secretory capacity ofsomatotrope cells in obesity: comparisonwith GH deficiency. International Journalof Obesity 21, 27–32.

Marcell, T.J., Wiswell, R.A., Hawkins, S.A.et al. (1999) Age-related blunting ofgrowth hormone secretion duringexercise may not be solely due toincreased somatostatin tone. Metabolism48(6), 665–670.

Merimee, T.J., Rabinowitz, D. & Fineberg,S.E. (1969) Arginine-initiated release of human growth hormone: factorsmodifying the response in normal man.New England Journal of Medicine 280,1434–1438.

Moretti, C., Favvri, A., Gnessi, L. et al.(1983) Naloxone inhibits exercise-induced release of PRL and GH inathletes. Clinical Endocrinology 18,135–138.

Mueller, E.E., Locatelli, V. & Cocchi, D.(1999) Neuroendocrine control ofgrowth hormone secretion. PhysiologicalReviews 79, 511–607.

Nindl, B.C., Kraemer, W.J., Marx, J.O. et al.(2003) Growth hormone molecularheterogeneity and exercise. Exercise andSport Sciences Reviews 31, 161–166.

Penelva, A., Pombo, M., Carballo, A. et al. (1993) Influence of sex, age andadrenergic pathways on the growthhormone response to GHRP-6. ClinicalEndocrinology 38, 87–91.

Pihoker, C., Middleton, R., Reynolds, G.A.,Bowers, C.Y. & Badger, T.M. (1995)Diagnostic studies with intravenous andintranasal growth hormone-releasingpeptide-2 in children of short stature.Journal of Clinical Endocrinology andMetabolism 80, 2987–2992.

Pincus, S.M., Gevers, E., Robinson, I.C.A.F.et al. (1996) Females secrete growthhormone with more process irregularitythan males in both human and rat.American Journal of Physiology.Endocrinology and Metabolism 270,E107–E115.

Popovic, V., Damjanovic, S., Micic, D. et al. (1995) Blocked growth hormone-releasing peptide (GHRP-6)-inducedGH secretion and absence of thesynergic action of GHRP-6 plus GH-releasing hormone in patients with

Thompson, D.L., Weltman, J.Y., Rogol,A.D. et al. (1993) Cholinergic and opioidinvolvement in release of growthhormone during exercise and recovery.Journal of Applied Physiology 75, 870–878.

Uusitupa, M., Siitonen, O., Harkonen, M. et al. (1982) Modification of the metabolicand hormonal response to physicalexercise by beta-blocking agents. Annals of Clinical Research 14 (suppl.),165–167.

Vahl, N., Jorgensen, J.O., Skjaerback, C. et al. (1997) Abdominal adiposity ratherthan age and sex predicts the mass andpatterned regularity of growth hormonesecretion in mid-life healthy adults.American Journal of Physiology 272,E1108–1116.

Veldhuis, J.D. (1995) The neuroendocrineregulation and implications of pulsatileGH secretion: gender effects.Endocrinologist 5, 198–213.

Veldhuis, J.D. (1996a) New modalities forunderstanding dynamic regulation ofthe somatotropic (GH) axis: explicationof gender differences in GHneuroregulation in the human. Journal of Pediatric Endocrinology and Metabolism9, 237–253.

Veldhuis, J.D. (1996b) Physiologicalregulation of growth hormone (GH)-insulin-like growth factor type I (IGF-I)axis: predominant impact of age,obesity, gonadal function, and sleep.Sleep 19, S221–S224.

Veldhuis, J.D. (1998) Neuroendocrinecontrol of pulsatile growth hormonerelease in man: relationship with gender.Growth Hormone and IGF Research 8,49–59.

Veldhuis, J.D. (2003) A tripeptidylensemble perspective of interactivecontrol of growth hormone secretion.Hormone Research 60, 86–101.

Veldhuis, J.D. & Bowers, C.Y. (2003a) Sex-steroid modulation of growthhormone (GH) secretory control: three-peptide ensemble regulation under dualfeedback restraint by GH and IGF-I.Endocrine 22(1), 25–40.

Veldhuis, J.D. & Bowers, C.Y. (2003b)Three-peptide control of pulsatile andentropic feedback-sensitive modes ofgrowth hormone secretion: modulationby estrogen and aromatizable androgen.Journal of Pediatric Endocrinology 16(suppl. 3), 587–605.

Veldhuis, J.D. & Yoshida, K. (2000) Impact of chronic training on pituitaryhormone secretion in the human. In:Sports Endocrinology (Warren, M.P. &

hypothalamopituitary disconnection:evidence that GHRP-6 main action isexerted at the hypothalamic level.Journal of Clinical Endocrinology andMetabolism 80, 942–947.

Pritzlaff, C.J., Wideman, L., Weltman, J.Y.et al. (1999) Impact of acute exerciseintensity on pulsatile growth hormonerelease in men. Journal of AppliedPhysiology 87, 498–504.

Pritzlaff, C.J., Wideman, L., Blumer, J. et al. (2000) Catecholamine release,growth hormone secretion, and energyexpenditure during exercise vs. recoveryin men. Journal of Applied Physiology 89,937–946.

Pritzlaff-Roy, C.J., Wideman, L., Weltman,J.Y. et al. (2002) Gender governs therelationship between exercise intensityand growth hormone release in youngadults. Journal of Applied Physiology 92,2053–2060.

Raynaud, J., Capderou, A., Martineaud, J.-P. et al. (1983) Intersubject variabilityof growth hormone time course duringdifferent types of work. Journal of AppliedPhysiology 55, 1682–1687.

Richmond, E.J., Rogol, A.D., Clark, W.,Dasdemir, D. & Veldhuis, J.D. (2001)Paradoxically heightened susceptibilityto GH autonegative feedback inmidpuberty compared with prepubertyin healthy boys. Paper presented at theEndocrine Society 83rd Meeting,Denver, CO.

Rudman, D., Kutner, M.H., Rogers, C.M. et al. (1981) Impaired growth hormonesecretion in the adult population. JournalClinical Investigation 67, 1361–1369.

Shuto, Y., Shibasaki, T., Otagiri, A. et al.(2002) Hypothalamic growth hormonesecretagogue receptor regulates growthhormone secretion, feeding, andadiposity. Journal Clinical Investigation109, 1429–1436.

Sutton, J. & Lazarus, L. (1974) Effect ofadrenergic blocking agents on growthhormone responses to physical exercise.Hormone and Metabolic Research 6,428–429.

Sutton, J. & Lazarus, L. (1976) Growthhormone in exercise: comparison ofphysiological and pharmacologicalstimuli. Journal of Applied Physiology 41,523–527.

Tanaka, K., Inoue, S., Numata, K. et al.(1990) Very-low-calorie diet-inducedweight reduction reverses impairedgrowth hormone secretion responses togrowth hormone-releasing hormone,arginine and l-dopa in obesity.Metabolism 39, 892–896.

Page 149: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

growth hormone and exercise 133

Constantini, N.W., eds.). Humana Press,Totowa, NJ: 57–76.

Veldhuis, J.D., Lassiter, A.B. & Johnson,M.L. (1990) Operating behavior of dualor multiple endocrine pulse generators.American Journal of Physiology.Endocrinology and Metabolism 259,E351–E361.

Veldhuis, J.D., Iranmenesh, A., Ho, K.K.Y.et al. (1991) Dual defects in pulsatilegrowth hormone secretion and clearancesubserve the hyposomatotropism ofobesity in man. Journal of ClinicalEndocrinology and Metabolism 72, 51–59.

Veldhuis, J.D., Liem, A.Y., South, S. et al.(1995) Differential impact of age, sexsteroid hormones, and obesity on basalversus pulsatile growth hormonesecretion in men as assessed in anultrasensitive chemiluminescence assay.Journal of Clinical Endocrinology andMetabolism 80, 3209–3222.

Veldhuis, J.D., Iranmanesh, A. & Weltman, A. (1997) Elements in thepathophysiology of diminished growthhormone (GH) secretion in aginghumans. Endocrine 7, 41–48.

Veldhuis, J.D., Iranmanesh, A., Mulligan,T. & Anderson, S. (2001) Interactiveregulation of postmenopausal growthhormone insulin-like growth factor axisby estrogen and growth hormone-releasing peptide-2. Endocrine 14, 45–62.

Veldhuis, J.D., Iranmanesh, A., Mulligan,T. & Bowers, C.T. (2002) Mechanisms of conjoint failure of the somatotropicand gonadal axes in ageing men.Novartis Foundation Symposium 242,98–118.

Weltman, A., Weltman, J.Y., Schurrer, R. et al. (1992) Endurance training amplifiesthe pulsatile release of growth hormone:effects of training intensity. Journal ofApplied Physiology 72, 2188–2196.

Weltman, A., Weltman, J.Y., Hartman,M.L. et al. (1994) Relationship between

Wideman, L., Weltman, J.Y., Shah, N. et al.(1999) The effects of gender on exercise-induced growth hormone release.Journal of Applied Physiology 87,1154–1162.

Wideman, L., Weltman, J.Y., Patrie, J.T. et al. (2000a) Synergy of l-arginine andgrowth hormone (GH)-releasingpeptide-2 (GHRP-2) stimulation of GHin men and women: Modulation byexercise. American Journal of Physiology.Regulatory, Integrative and ComparativePhysiology 279, R1467–R1477.

Wideman, L., Weltman, J.Y., Patrie, J.T. et al. (2000b) Synergy of l-arginine andgrowth hormone (GH)-releasingpeptide-2 on GH release: influence ofgender. American Journal of Physiology.Regulatory, Integrative and ComparativePhysiology 279, R1455–R1466.

Williams, T., Berelowitz, M., Joffe, S.N. et al. (1984) Impaired growth hormoneresponses to growth hormone-releasingfactor in obesity: a pituitary defectreversed with weight reduction. New England Journal of Medicine 311,1403–1407.

Winer, L.M., Shaw, M.A., Baumann, G.(1990) Basal plasma growth hormonelevels in man: new evidence forrhythmicity of growth hormonesecretion. Journal of Clinical Endocrinology and Metabolism 70,1678–1686.

Zaccaria, M., Vanier, M., Piazza, P.,Noventa, D. & Ermolao, A. et al. (1999)Blunted growth hormone response to maximal exercise in middle-agedversus young subjects and no effect ofendurance training. Journal of ClinicalEndocrinology and Metabolism 84,2303–2307.

Zadik, Z., Chalew, S.A., McCarter, R.J. et al.(1985) The influence of age on the 24-hour integrated concentration of growthhormone in normal individuals. Journalof Clinical Endocrinology and Metabolism60, 513–516.

age, percentage body fat, fitness, and 24-hour growth hormone release inhealthy young adults: effects of gender.Journal of Clinical Endocrinology andMetabolism 78, 543–548.

Weltman, A., Weltman, J.Y., Womack, C.J.et al. (1997) Exercise training decreasesthe growth hormone (GH) response toacute constant-load exercise. Medicineand Science in Sports and Exercise 29,669–676.

Weltman, A., Pritzlaff, C.J., Wideman, L. et al. (2000a) Exercise-dependent growthhormone release is linked to markers of heightened central adrenergicoutflow. Journal of Applied Physiology89, 629–635.

Weltman, A., Pritzlaff, C.J., Wideman, L. et al. (2000b) The relationship betweenexercise intensity and growth hormone(GH) release is attenuated in older men. Paper presented at the FourthInternational Conference of the GrowthHormone Research Society, Gothenberg,Sweden.

Weltman, A., Anderson, S.M., Wideman,L. et al. (2001) Impact of short-termestrogen supplementation inpostmenopausal women onspontaneous and exercise stimulatedpulsatile growth hormone (GH)secretion. Paper presented at the 83rdAnnual Meeting of the EndocrineSociety. Denver, CO.

Weltman, A., Brill, K., Weltman, J.Y. et al.(2002) GHRP-2 partially rescuesimpaired exercise stimulation of growthhormone (GH) release in older men.Paper presented at the 84th AnnualMeeting of the Endocrine Society, SanFransisco, CA.

Weltman, J.Y., Frick, K., Watson, D. et al.(2002) Comparison of continuous andintermittent exercise on 24-h growthhormone secretion in obese and non-obese young men. Paper presented at the 84th Annual Meeting of theEndocrine Society, San Fransisco, CA.

Page 150: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

134

Introduction

The pituitary proopiomelanocortin (POMC) sys-tem is activated under physical, psychological andimmunological stressors, which induce the releaseof POMC fragments into the cardiovascular com-partment. The response to physical stress includesthe release of a variety of neuronal or humoral sig-nals from nervous, endocrine or immune systemsending up with morphological or functional altera-tions, for example of muscular tissue or of the car-diovascular system (Teschemacher 2003).

Adrenocorticotropic hormone (ACTH) and β-endorphin are the compounds mostly studied underphysical stress conditions, whereas further POMCfragments, such as β-lipotropin (β-LPH), are alsoreleased upon physical exercise. The secretion ofPOMC derivatives during exercise is an adaptiveattempt of the athlete’s organism to cope with dif-ferent stress situations. It is intimately linked to avariety of psychological strategies that facilitate theorganism’s navigation through a stressful envir-onment (McCubbin 1993). ACTH and β-endorphinappear to be released during exercise in a sufficientintensity and duration. The exercise response mayalso be affected by the training status of the indivi-dual and the population being investigated (Goldfarb& Jamurtas 1997). Aerobic activities (enduranceexercise) are quite different from anaerobic activit-ies, such as resistance exercise. In case of situationsof overtraining challenge, a reduced ACTH responsemay reflect the athlete’s impaired ability to copewith the stress situation. Whereas short-term over-training (overreaching) can be reversed by a moreprolonged period of recovery, further exposure to

stressors induces overtraining syndrome. Changesin circulating hormonal concentrations have beenproposed as indicators of overtraining, althoughsuch hormonal changes are not always observed(Fry et al. 1998).

However, it remains to be elucidated, which purpose for POMC fragments are released into thecardiovascular compartment related to exerciseconditions. This has to be emphasized despite of afrequently offered answer suggesting the oppositeby referring to an ‘adaptation to stress’. The clari-fication of the functional significance of the POMCsystem, which is obviously activated by the humanorganism every day and under multiple stress con-ditions, still remains a target of interest (Teschemacher2003). The exact mechanism(s) responsible for exer-cise-induced increases in β-endorphin, ACTH andcortisol remain speculative. They may also differ independence of the duration of exercise stress or independence of the exercise stress intensity (i.e. sub-maximal and maximal) exercise stress, when high-intensity exercise at supramaximal oxygen (O2)consumption (Vo2max) does not longer produce sig-nificant increase in plasma β-endorphin (Kraemer,W.J. et al. 1993). This review summarizes the currentknowledge about POMC in exercise physiology.

POMC and its occurrence in the history of life

POMC—phylogenetic ontogeny and POMC-derived peptides

Proopiomelanocortin (POMC) is older than 500 mil-lion years. This protein has not only been found in

Chapter 11

Proopiomelanocortin and Exercise

HEINZ W. HARBACH AND GUNTER HEMPELMANN

Page 151: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 135

mammalians but already in invertebrates (Denef &Van Bael 1998), and is also expressed in unicellularorganisms (Salzet et al. 1997). So far, POMC might beof significance both for the primitive living beingsand for the highest developed living systems. Muchabout the ontogeny of the human POMC system isnot known, but it is known that POMC fragmentsare prepared in the human fetal organism alreadyfrom the 5th week of gestation on (Facchinetti et al.1987). POMC and two other major prohormones,proenkephalin and prodynorphin, give rise to thethree ‘families’ of opioid peptides: the endorphin,the enkephalin and the dynorphin families.

Peptide hormones and neuropeptides are synthes-ized as part of a large protein precursor moleculePOMC from which they are post-translationally lib-erated and modified through enzymatic processing(Fig. 11.1). In humans, there is one POMC gene perhaploid genome located on chromosome 2 (p23). Itis 7665 base pairs long and consists of three exonsseparated by two large introns (3–4 and 2–3 kbrespectively). The first exon is about 100 nucleotidesin length and contains the 5’-untranslated region of the POMC mRNa. Exon 2 consists of about 150nucleotides and contains, besides a small portion

of the 5’-untranslated sequence, the sequence of the signal peptide and the first 18 amino acids ofPOMC. The third exon encodes the rest of thePOMC precursor with the sequences of ACTH, β-LPH and β-endorphin. Biological active peptidesarising from POMC are subjects to further process-ing, thus yielding smaller peptide products withdistinct biological activity. So far, POMC is pro-cessed to pro-γ-melanocyte stimulating hormone(pro-γ-MSH) (also called N-POMC), corticotropin-like-intermediate protein (CLIP), joining peptide(JP), ACTH and β-LPH (Fig. 11.1). There are at least10 β-endorphin derivatives known to exist in themammalian organism, β-endorphin (1–27), (1–26),(1–17) and (1–16), as well as their N-acetylatedforms, and in addition N-acetyl- β-endorphin (1–31)(Teschemacher et al. 1990a; Höllt 1993; Young et al.1993). Pro-γ-MSH is cleaved to give an N-terminalpeptide (N-POMC (1–49)) ACTH to give α-MSHand CLIP, and β-LPH is cleaved to β-endorphin andγ-LPH. The JP has no biological activity, although it contains a single cysteine residue, unique inhumans, which allows it to dimerize, and it isthought that POMC may also dimerize within the cell during biosynthesis (Bicknell et al. 1996).

Signal peptide

ACTH β-LPH

β-END

β-END

γ-LPH

β-END (1–31)

β-END (1–27)

β-END (1–26)

β-END (1–17)

β-END (1–16)

acetyl-N-

acetyl-N-

acetyl-N-

acetyl-N-

acetyl-N-

β-MSHCLIPN-term fragment JPγ-MSH α-MSH

Fig. 11.1 Schematic representation of known products of enzymatic processing of proopiomelanocortin (POMC). Signalpeptide; N-terminal fragment; adrenocorticotropic hormone (ACTH); β-lipotropin (LPH); γ-LPH; α-melanotropin (MSH); β-MSH; γ-MSH; corticotropin-like intermediate lobe peptide (CLIP); joining peptide (JP); β-endorphin (1–31) (β-END (1–31)); β-END (1–27); β-END (1–26); β-END (1–17); β-END (1–16); acetyl-N-β-END (1–31); acetyl-N-β-END(1–27); acetyl-N-β-END (1–26); acetyl-N-β-END (1–17); acetyl-N-β-END (1–16).

Page 152: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

136 chapter 11

N-POMC is involved in adrenal mitogenesis and α-MSH controls pigmentation in lower vertebrates.

The mRNA in the pituitary is 1072 bases long,whereas the transcript in the brain is longer andmost of the transcripts in the periphery are shorter,i.e. about 800 bases long. POMC is produced in particularly high amounts in the pituitary gland.The POMC mRNA in the pituitary, in the brain andin the periphery as far as comparable with pituitarymRNA length is translated into the amino acidsequence of ‘pre-proopiomelanocortin’, ‘pre-POMC’.After cleavage of the N-terminally located signalsequence from pre-POMC, POMC is left, which is a protein of 241 amino acid residues, from whichPOMC fragments such as ACTH or β-endorphin arereleased (for review see Höllt 1993; Bertagna 1994).

POMC localization in the human organism

The main site of expression of the POMC gene is thepituitary gland, in particular the corticotropic cellsof the anterior lobe and the melanotropic cells of theintermediate lobe. The intermediate lobe as the genu-ine location of the melanotropic cells in lowerspecies is vestigial in the human pituitary gland. So far the human organism lacks a well-definedintermediate lobe containing melanotropic cells.However, the POMC-expressing cells in the anteriorlobe are able to process POMC according to eithertype of enzymatic cleavage pattern, the melanotro-pic as well as the corticotropic one. These enzymaticsystems, however, apparently are still regulatedseparately (Evans, V.R. et al. 1994).

Outside the pituitary POMC transcripts havebeen found as well, for example in the arcuatenucleus of the hypothalamus and other regions ofthe brain. Moreover, POMC-like transcripts havebeen detected in a number of peripheral tissues.These are the thyroid gland, thymus, adrenal med-ulla, gonads, placenta, pancreas, kidneys, spleen,liver, gastrointestinal wall, skin, monocytes, macro-phages and T-cells.

POMC expression in the human organism

In the corticotropic cells POMC is glycosylated andphosphorylated and subsequently enzymatically

cleaved into three big fragments, a so-called 16 K-fragment, ACTH and β-LPH (Fig. 11.1); the latterPOMC fragment consists of the sequences of γ-LPH and β-endorphin, which are released in smallamounts also. In the melanotropic cells, the samePOMC fragments are released from POMC as in the corticotropic cells; however, they are furtherprocessed to release a series of smaller frag-ments, which are in part N-terminally acetylated orC-terminally amidated. α-Melanocyte stimulatinghormone (α-MSH) or acetylated β-endorphin frag-ments are typical examples for this type of POMCderivatives (Loh 1992; Young et al. 1993; Castro &Morrison 1997).

POMC—the precursor of ACTH and ββ-endorphin—and the endocrineregulation of the response to exercise stress

POMC release under physical stress conditions

The ‘corticotropic part’ of the POMC system is activated under the conditions of physical exercise,which results in the release of ACTH and β-endorphin immunoreactive material (IRM). ACTHwas studied as the main representative of thePOMC system for a long time under the aspect of stress-induced activation of the ‘hypothalamic–pituitary–adrenal axis’ (HPA axis) (Ganong et al.1987; Tache & Rivier 1993). ACTH is regarded as amain valid parameter of the endocrine stressresponse besides epinephrine, norepinephrine andcortisol (Adams & Hempelmann 1991). Later β-endorphin and β-LPH were recognized to be re-leased from the pituitary under identical or similarstress conditions (Owens & Smith 1987; McLoughlinet al. 1993).

ACTH and β-endorphin IRM concentrations inthe plasma mostly were secreted in equimolar quantities in response to exercise (Akil et al. 1984; DeMeirleir et al. 1986; Farrell et al. 1987; Rahkila et al.1988; Strassman et al. 1989; Schwarz & Kindermann1990; Heitkamp et al. 1993; Schulz et al. 2000).However, as shown in female marathon runners,basal levels of ACTH and β-endorphin IRM can bedifferent and the increase of ACTH levels under

Page 153: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 137

certain conditions, for example upon running toexhaustion, can exceed the increase of β-endorphinIRM by a factor of five (Heitkamp et al. 1996).

Acute stress can be defined biochemically bymeasuring an increase in catecholamine release.Chronic stress has proved more difficult to define inbiochemical terms, although in psychological termsit has been considered as the lack of ability to copewith the environment or loss of control in a long-term situation (McLoughlin et al. 1993). In the case ofathlete’s feeling subjective symptoms of stress, com-bined with accelerated fatigability or diminishedphysical performance, a state of overtraining stressis given.

POMC response to acute aerobic and endurance exercise

Assuming that the stress response is a neuroen-docrine mechanism that occurs in anticipation ofphysical exercise, De Vries et al. (2000) investigatedwhether an incremental exercise protocol could beused as a model stressor. Subjects cycled at 40–100%of the power output at the maximal O2 consumption(Vo2max) each up to exhaustion. Their results showedthat increases in heart rate, lactate, epinephrine and norepinephrine reflect the relative workload, incontrast to increases in ACTH hormone and β-endorphin, which were observed only after exercisereached an intensity of 80% Vo2max. Their resultsdemonstrated that activation of stress hormonesoccur at different time points, the delayed responseof the HPA axis during incremental exercise con-trasted with the non-delayed HPA axis responseobserved during psychological stress (De Vries et al.2000). Farrell et al. (1983) compared ACTH levelsbefore and after submaximal (80% of maximal O2consumption [Vo2max]) and exhaustive isotonicexercise (100% Vo2max) and also concluded thatexercise-induced increases in plasma ACTH andtheir correlation with circulating cortisol depend onthe intensity of isotonic exercise. At workloads of40–60% Vo2max β-endorphin was not significantlyelevated, but it was significantly elevated at 80%Vo2max (Donevan & Andrew 1987; Langenfeld et al.1987; McMurray et al. 1987; Sforzo 1989). In a studyby Duclos et al. (1997) ACTH levels after running

exercise proved to be elevated in marathon-trainedathletes in comparison with untrained volunteers.However, since cortisol levels did not reflect thisdifference, the authors of this study hypothesized adecreased HPA axis sensitivity to cortisol negativefeedback. Results of a following study from Ducloset al. (1998) discarded this hypothesis of a decreasedadrenal sensitivity of the ACTH targets after pitui-tary adrenal stimulation.

Marquet et al. (1999) administered low and highdoses of dexamethasone. After the last dose the subjects performed a maximal cycling exercise, and blood was sampled just before and after eachexercise bout. Blood levels of ACTH, β-endorphin,cortisol and sex steroids except testosterone werelowered by dexamethasone at rest and after exer-cise. These effects were interpreted as an impair-ment of the adaptation to intense physical loads.

Controversy exists regarding the effects of duration of exercise and exercise dosage on β-endorphin release during exercise. Angelopoulos(2001) demonstrated an increased concentration ofβ-endorphin maintained over time during intenseexercise (trials at 80% Vo2max for 10–30 minutes),whereas Goldfarb et al. (1991) reported a gradualincrease in β-endorphin concentration over time at80% Vo2max, and Taylor et al. (1994) observed a linear increase in β-endorphin concentration withtime. The disparity among the results may be due to differences in the exercise dosage or a matchbetween the rate of β-endorphin release and dis-appearance at 50% Vo2max conditions. The secretionof β-endorphin and ACTH were also described independence of the intensity of exercise (Rahkila et al.1987, 1988). However, results from Petraglia et al.(1990) at marathon race conditions and submaximalexercise at 50% Vo2max indicate that the duration butnot the workload were responsible for β-endorphinand β-LPH release.

Goldfarb and Jamurtas (1997) reviewed signifi-cant increases in β-endorphin at workloads of 60%and 90% Vo2max only at the higher workload anddependant on the duration of exercise, respectively.Graded or incremental exercise of an aerobic natureappears to increase β-endorphin. Exercise at 66%and 57% Vo2max was reported to increase plasma β-endorphin in the untrained and trained individuals,

Page 154: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

138 chapter 11

respectively. The authors indicated that the β-endorphin levels did not differ significantly betweenthe endurance-trained and untrained participantseither prior to or during exercise.

Lac et al. (1999) present reactions of cortisol to adexamethasone treatment via ACTH blockade andto an exercise bout at Vo2max. After exercise, plasmaACTH rose to 600% of basal value, whereas cortisolwas unaffected, which was explained by differencesin peripheral metabolism.

Changes of plasma ACTH and β-endorphin wereobserved under high-intensity cycle exercise andexposure to a simulated altitude of 4300 m (hypo-baric and hypoxic) and under sea level conditions:Exercise at acute hypobaric hypoxia elicited no sig-nificantly different responses in plasma β-endorphin,ACTH or cortisol and no changes in β-endorphin/ACTH molar ratio than those elicited under nor-mobaric conditions (Kraemer, W.J. et al. 1991). Asignificant increase in serum ACTH was observedin response to a 21 km non-competitive race at lowaltitude (350 m below sea level) in comparison to620 m above sea level; it was proposed that ACTHmay play a role in acclimatization to exercise at lowaltitudes (el Migdadi et al. 1996).

Repeated physical exercise including the follow-ing recovery period may be a useful model to studythe effect of various rest intervals on subsequentstress reactions. Therefore, Ronsen et al. (2002)designed a study that compared neuroendocrineand immune responses during days with two equalbouts of high-intensity endurance exercise, but dif-ferent periods of rest between the first and secondbout. They demonstrated that when a second boutof exercise was performed after 3 h compared with 6 h of rest, increases in epinephrine, ACTH and cortisol were augmented. They showed that recoverytime as such is a significant factor in achievinghomeostasis between repeated sessions of endur-ance exercise.

β-Endorphin and ventilatory responsiveness. To invest-igate the hypothesis that endurance exercise maylead to a decrease in ventilatory chemosensitivity aspossibly mediated by an increase in endogenous β-endorphin, the hypercapnic ventilatory responsive-ness and circulating β-endorphin immunoreactivity

was measured at marathon race. All runners experi-enced a rise in β-endorphin activity from pre-marathon to immediate post-marathon. However,hypercapnic ventilatory responsiveness showed nosignificant change. Mahler et al. (1989) concludedthat the natural increase in endogenous β-endorphinactivity associated with marathon running does not modulate central chemosensitivity. Adult malestudents were given Vo2max tests to confirm theiraerobic fitness levels. The ‘fit group’ showed sig-nificantly lower heart rate responses than the ‘non-fit group’, and these fitness group differences wereabolished by the opioid antagonist naltrexone.McCubbin (1993) suggested that regular aerobicexercise conditioning augments release of inhibit-ory opioids.

POMC response to acute resistance exercise

Aerobic activities (endurance exercise) are quite dif-ferent from anaerobic activities, such as resistanceexercise. A decrease in β-endorphin plasma concen-tration following resistance exercise was observedas compared with pre-exercise (Pierce et al. 1994),and in addition, no relationship between affect andβ-endorphin response to exercise was observed(McGowan et al. 1993). As Goldfarb and Jamurtas(1997) also described, it appeared there is no definit-ive response of β-endorphin to resistance exercise.

In heavy-resistance exercise, it appeared that theduration of exercise, length of the rest periodsbetween exercise sets and blood lactate were keyexercise variables that influence increases in plasmaβ-endorphin concentration (Kraemer, W.J. et al.1993). Their dataaonly one of six heavy-resistanceexercise protocols examined resulted in a significantincrease in plasma β-endorphin and serum cortisolconcentrationsademonstrated that the exercise sti-mulus for β-endorphin increase was characterizedby longer-duration sets and shorter rest periodsbetween sets and exercises. The number of repeti-tions that the resistance (i.e. intensity) allows in theset (i.e. duration) and the rest period length are theprimary determinants of the physiological stress(Kraemer, W.J. et al. 1989b). β-Endorphin increasedat the end of resistance exercise during energy bal-ance, but significantly only during negative energy

Page 155: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 139

balance (Walberg-Rankin et al. 1992). β-Endorphinor whole blood lactate were not influenced by re-sistance training experience in elite weightlifters(Kraemer, W.J. et al. 1992). It appears that there is no definitive response of β-endorphin to resist-ance exercise. Non-exhaustive performance such asheavy resistance exercise, although requiring highmuscular activity for short time periods, did not leadto an increase of β-endorphin levels at all (Pierce et al. 1993b), even decreased them (Pierce et al. 1994),or only allowed an increase under approximatelyexhaustive conditions (Kraemer, W.J. et al. 1993).Schulz et al. (2000) demonstrated the increase ofACTH and β-endorphin IRM concentrations in theplasma upon anaerobic exercise, correlated with theincrease of lactate levels observed upon anaerobicexercise. Authentic β-endorphin (1–31) was onlyfound in two plasma samples containing minor con-centrations of the peptide. They concluded that the β-endorphin IRM released into blood underanaerobic exercise was identical with authentic β-endorphin (1–31) only to a minor extent. The majorpart of the material in fact released into the bloodupon anaerobic exercise was probably identicalwith β-LPH.

The results examining resistance exercise and β-endorphin response are equivocal: partly due tothe selection of participants; partly due to the inten-sity, duration and rest periods of exercise utilized(Kraemer, W.J. et al. 1989; Goldfarb & Jamurtas1997); and partly due to the specificity of assays utilized (Harbach et al. 2000; Schulz et al. 2000).

Ethanol did not increase circulating cortisol con-centration above that caused by the resistance exer-cise, but it appeared to have a more prolonged effectwithout a change in circulating ACTH (Koziris et al.2000). Unchanged resting concentrations of cortisolduring short periods of heavy strength trainingwere also observed by Fry et al. (1998) and Raastad et al. (2001).

Chronic training effects and POMC response:chronic aerobic and endurance exercise versuschronic resistance exercise

POMC response to chronic aerobic and endurance exercise. Buono et al. (1987) observed a significant

increase in Vo2max of volunteers but a bluntedACTH response to an absolute submaximal workrate in a 12-week training program. In addition,Luger et al. (1987) observed decreased ACTH levelsin trained subjects under consideration of absoluteworkload. In contrast, Ronkainen et al. (1986), whoinvestigated the effects of endurance exercise on the function of adrenal cortex of female runners,supposed no alteration of the function of the adrenalcortex by chronic endurance exercise, when res-ponses of cortisol to intravenous ACTH injectionswere estimated.

POMC response to chronic resistance exercise. Datafrom Kraemer, W.J. et al. (1992) on effects of resist-ance training experience demonstrated an increaseof β-endorphin, testosterone and lactate, whereasonly testosterone was influenced by 2-years’ train-ing experience.

Deschenes et al. (2002) tested whether muscleunloading were attributable to adaptations of theneural system rather than modifications of myofibersize and confirmed that the loss of muscular strengthcould primarily be attributed to a decreased capa-city of the nervous system to excite the muscle. In contrast to resistance training, which results instrength gains and muscle hypertrophy, muscleunloading evokes reductions in muscle perform-ance. In addition, they observed that muscle un-loading affects the hormonal milieu in a mannerthat promotes muscle atrophy: increase of thecatabolic hormone cortisol in the absence of a con-comitant elevation of ACTH.

Sleep deprivation did not alter the resting β-endorphin response, nor did it affect the β-endor-phin response to high-intensity exercise (McMurrayet al. 1988).

Influence of training. The influence of training onbasal plasma levels or on levels under exercise con-ditions has been reviewed (Goldfarb & Jamurtas1997). A number of studies differing in the composi-tion of volunteer groups as well as using differentprotocols obviously revealed controversial results:Training has been reported to have increased, madeno difference to or decreased β-endorphin levels following exercise. Goldfarb and Jamurtas (1997)

Page 156: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

140 chapter 11

interpreted the discrepancy in the literature as inpart related to the type of training and its intensity,the methods used to measure β-endorphin and themode of exercise utilized. A clear-cut experimentaldesign was presented by Engfred et al. (1994), whichdid not just compare trained and untrained volun-teers but looked at the same subjects in a state beforeand after training as conducted and controlled inthe frame of their own study. Training-inducedchanges in response to exercise of norepinephrine,epinephrine, growth hormone, β-endorphin andinsulin were similar in the groups. Incrementalexhausting and prolonged exhausting enduranceexercise such as marathon running induced anincrease of similar magnitude in both β-endorphinand ACTH concentration (Heitkamp et al. 1993).ACTH and β-endorphin response to exercise work-load proved to be dramatically reduced after a 5weeks’ training protocol. In a similarly designedstudy, untrained females were subjected to 8 weeks’endurance training and the effects of running 30 minutes three times a week on ACTH and β-endorphin levels were measured (Heitkamp et al.1998). Basal β-endorphin levels were not altered bythe training program, but basal ACTH levelsincreased. Both, ACTH as well as β-endorphin levels, were dramatically increased immediatelyafter the exercise; this increase, however, was foundto be attenuated in case of β-endorphin. Thus, train-ing appeared to provoke a POMC fragment-specificresponse, which was difficult to recognize just uponacute exercise challenge of the POMC system. Resultsobtained with untrained and trained volunteers also showed differences between the two groupsconcerning different ACTH and β-endorphin res-ponses, which can be interpreted in the same way(Diego Acosta et al. 2001). Training also appears toinfluence β-endorphin metabolism in the restingstate and during exercise in dependence of trainingpreviously performed; mild endurance exercise50% Vo2max resulted in no change of β-endorphin,whereas exercise at 66% was reported to increase β-endorphin (Viru & Tendzegolskis 1995).

Luger et al. (1987) examined plasma ACTH, cortisol and lactate responses in sedentary subjects,moderately trained and highly trained runners. Basalconcentrations of plasma ACTH were elevated in

the highly trained subjects. Like the levels of plasmaACTH and cortisol, the level of plasma lactate wascoupled to exercise intensity, since all groups hadsimilar plasma lactate responses at matched exer-cise intensities. Trained subjects required muchhigher absolute workloads to produce comparableelevations in plasma lactate concentrations; theyhad much less activation of the HPA axis as com-pared with untrained subjects at matched absoluteworkloads. Daily strenuous exercise was inter-preted to lead to chronic ACTH hypersecretion and adrenal hyperfunctionaphysical conditioningassociated with adaptation mechanisms such as theability to increase the capacity to handle a higherworkload with less pituitary–adrenal activation(Luger et al. 1987). Data of the same group showingelevated basal concentrations of ACTH and cortisoland a blunted response to exogenous corticotropin-releasing hormone (CRH) were compatible withmild sustained hypercortisolism in highly trainedrunners (Luger et al. 1988).

In studies comparing sedentary with trained subjects, no training-induced adaptation of POMCderivative release was observed under differentexercise conditions (Kraemer, R.R. et al. 1989;Goldfarb et al. 1991). Three training groups wereelected for maximal treadmill exercise. Sprint inter-vals, endurance training and combination trainingwere observed over 10 weeks. No training-inducedhormonal changes were observed for the endu-rance group. While exercise-induced increases wereobserved, the combination group exhibited signifi-cant post-training reductions in plasma responses ofβ-endorphin and ACTH (Kraemer, W.J. et al. 1989a).

In comparison with men, female marathon run-ners showed higher baseline concentrations, lesserincreases in β-endorphin, lower baseline concentra-tions and larger increases in ACTH concentrationafter marathon running and treadmill (Heitkamp et al. 1996). In addition, the same group observed a tendency to elevated basal ACTH in endurancetraining (Heitkamp et al. 1998).

POMC release under extreme physical stress conditions.Ultramarathon foot race is extreme physical stress;resting serum ACTH and β-endorphin levels weresignificantly elevated above normal range (altered

Page 157: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 141

baseline hormonal state), ACTH remained elevated,and β-endorphin plasma concentration was withinthe normal range without significant change andwas interpreted as a hormonal adaptation to pro-longed stress (Pestell et al. 1989). A more pronouncedresponse in trained subjects was only seen underextreme exercise conditions (Farrell et al. 1987).

Tharp already in 1975 made the following state-ments concerning the role of glucocorticoids in exer-cise and training: Exhaustion produces a decrease in plasma glucocorticoid, which may represent adefense mechanism to prevent depletion of bodyresources. Chronic exercise training producesadrenal cortex hypertrophy and usually a smallerrise in plasma glucocorticoids during an acute exer-cise bout than that obtained with non-trained sub-jects. The changes in glucocorticoid response duringtraining appear to be produced by decreasedresponsiveness to the adrenal cortex itself to ACTHstimulation and possibly by adaptation of the HPAaxis which reduces the ACTH released in responseto stress (Tharp 1975).

Data from Viru and Tendzegolskis (1995) demon-strated that levels of γ1–17-endorphin and α1–16-endorphin and the ratios to β-endorphin weresignificantly higher in untrained individuals, indi-cating an alteration of the metabolism of β-endorphinin the resting state and during exercise as depen-dent on previous training.

Overtraining and addiction to exercise

This chapter refers to exercise physiology in a stateof imbalance between strain of exercise training and the athlete’s tolerance of stress which leads toovertraining. The literature on overtraining is con-fusing because of a lack of universal terminology(Fry & Kraemer 1997). Overtraining can be definedas any training performed with incomplete recoverybetween bouts of exercise, leading to physical per-formance decrements (Fry & Kraemer 1997; Raastadet al. 2001). A further lack in the literature is thatstudies of overtraining do not consequently utilizeadequate changes in exercise volume or intensity, or do not carefully consider rest and recovery characteristics of the training programme, or do notdeal with actual decreases in performance (Fry &

Kraemer 1997). Whereas short-term overtraining(overreaching) can be reversed by a more prolongedperiod of recovery, further exposure to stressorsinduces overtraining syndrome. Substrates such aslactate, urea, enzymes (e.g. creatine kinase) or hor-mones (e.g. cortisol, testosterone, growth hormone)are of interest in overtraining research, but we willonly report on ACTH and β-endorphin as represen-tative POMC derivatives in the blood in overtrain-ing analysis.

Urhausen et al. (1995) in parallel with Barron et al.(1985) and Fry and Kraemer (1997) (for review seeUrhausen et al. 1995; Fry & Kraemer 1997) observedan impaired exercise-induced increase in ACTHafter exhausting short-endurance test at 110% of the individual threshold or in response to insulin-induced hypoglycemia in overtrained marathonrunners, which was ascribed to a negative feedbackthrough cortisol, depletion of the pituitary ACTHpool or change of the intracellular homeostasis(Urhausen et al. 1995). Kraemer et al. (1989b) alsoreported an impaired exercise-induced rise of ACTHin athletes during overtraining. Repeated acute orchronic exposure to a particular stress results inadaptation, whereby the HPA axis becomes lessresponsive to subsequent or continued exposure to that particular stress. Data from Wittert et al.(1996) show that intense physical training leads toadaptive changes in basal HPA function, includ-ing a phase shift and increased pituitary in basalHPA function, a phase shift and increased pituit-ary ACTH secretion, but also a blunted cortisolresponse.

During heavy endurance training or overreach-ing periods, the majority of findings give evidenceof a reduced adrenal responsiveness to ACTH, com-pensated by an increased pituitary ACTH release(Lehmann et al. 1998). A decreased β-endorphinresponse was also reported in overtrained athletes(Urhausen et al. 1995). There is additionally evid-ence for decreased intrinsic sympathetic activityand sensitivity of target organs to catecholamines(Lehmann et al. 1998).

The parasympathetic, Addison type representsthe dominant modern type of the overtraining syn-drome. In an early stage, despite increased pituitaryACTH release, the decreased adrenal responsiveness

Page 158: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

142 chapter 11

is no longer compensated and the cortisol responsedecreases. In an advanced stage of overtraining syn-drome, the pituitary ACTH release also decreases.However, this complete pattern is only observedsubsequent to high-volume endurance overtrainingat high caloric demands. The functional alterationsof pituitary–adrenal axis and sympathetic systemcan explain persistent performance incompetence inaffected athletes (Lehmann et al. 1998).

An exercise effect to be rather elicited in the central nervous system than in the periphery isaddiction to exercise. However, in a study targetingthis question, scores on exercise-dependence surveywere not correlated with β-endorphin plasma levels(Pierce et al. 1993a).

Neuroendocrine responses to high volume resis-tance exercise overtraining can be grouped withhighly aerobic activities, whereas excessive resis-tance training intensity (anaerobic activity) producesa distinctly different neuroendocrine profile (Fry &Kraemer 1997). Cortisol concentrations in responseto resistance exercise at increased training volumeor increased training intensity were compared:Cortisol increased at rest and acute increased train-ing volume, whereas no change or a slight decreasewas observed at increased training intensity. How-ever, there was a lack of increased cortisol levelswith high-intensity resistance exercise overtraining.This might be of interest for overtraining research,since it has been shown that increased circulatinglevels of cortisol may be associated with psycholo-gical depression (Fry & Kraemer 1997).

It appears quite different when anaerobic activit-ies, such as resistance exercise, are compared withaerobic activities. Many of the overtraining symp-toms identified for aerobic exercise have not beenreported for anaerobic overtraining protocols, andaltering some of the acute training variables forresistance exercise results in a variety of differentphysiological responses (Fry et al. 1998). Exercise-induced cortisol levels decreased when training volume was doubled, arguing that the presence of increased resting levels of cortisol contributes an exhaustion of the HPA axis, thus preventing anadequate cortisol response to acute stress. In thepresence of maximal intensity resistance exerciseovertraining, no changes were observed for resting

cortisol levels. Such responses are contrary to highvolume resistance exercise and overtraining withhighly aerobic activities. It becomes apparent thatsome of the classical signs of overtraining, based on data from endurance athletes, cannot necessarilybe applied to overtraining resulting from highlyanaerobic activities (Fry & Kraemer 1997).

With ACTH and resistance exercise overtraining,acute ACTH levels are attenuated with increasedexercise intensity. When strength performance hasbeen decreased via maximal intensity resistanceexercise overtraining, circulating ACTH levels didnot change either at rest or after exercise (Fry &Kraemer 1997).

Previous research has attempted to identifyendocrine markers (‘overtraining markers’) of animpending or concurrent overtraining syndromefor both aerobic and anaerobic activities. Data fromMcGowan et al. (1993) support research showingresistance exercise does not produce the significantincrease in β-endorphin immunoreactivity widelyreported after endurance exercise. In addition, they showed no relationship between affect (mood)and β-endorphin response to resistance exercise.Despite speculations on an essential participation ofhypothalamic–pituitary hormonal changes in thepathogenesis of overtraining, only few results withrespect to athletes who were actually in a state ofovertraining are available (Urhausen et al. 1995).Training intensity effect monitoring for resistanceexercise was only addressed by Fry and colleagues(Fry & Kraemer 1997; Fry et al. 1998). Because of the lack of association between endocrine and per-formance alterations, they argued that the type ofexercise does not readily permit use of hormonalalterations and endocrine adaptations, respectively,to monitor impending overtraining. In addition, it appeared that short-term, high-relative-inten-sity resistance exercise overtraining may not be successfully monitored via circulating hormonal concentrations.

Implications of these neuroendocrine responsesto overtraining for adequate training should bemanaged by dosing the training stresses in a vari-able manner to avoid overtraining with long-termdisruption of homeostasis (Fry & Kraemer 1997).The magnitude of temporary fatigue and recovery

Page 159: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 143

rate (after heavy resistance exercise) may be an indica-tion of effectiveness for long-term adaptations of the neuromuscular system, which make it necessaryto start the next training session after completerecovery (Ahtiainen et al. 2003).

Cardiovascular effects

Exercise can be regarded as one of many stressorsprovoking cardiovascular reactions of the organismand therefore activating the pituitary POMC sys-tem. Shen et al. (1992) observed an exercise-inducedrise of β-endorphin plasma concentrations also inthe presence of naloxone. Their results confirm arise in intrinsic heart rate (heart rate following auto-nomic blockade) and β-endorphin concentrationswith acute exercise, but indicate that the changes inintrinsic heart rate are not β-endorphin related.

The β-endorphin response to exercise is of specialinterest in patients with coronary artery disease(CAD). Letizia et al. (1996) observed patients withsuspected CAD and those with definite CAD undercycloergometric stress tests. At peak exerciseplasma levels of β-endorphin and ACTH increasedconcomitantly in subjects that did not exhibit clinicand electrocardiogram (ECG) signs of ischemia during stress test. β-Endorphin, ACTH and cortisolincreased further during recovery. β-Endorphinsignificantly increased at peak exercise in patientswith CAD and negative stress test, but ACTH andcortisol plasma levels were not significantly modi-fied. On the contrary in those patients with a posit-ive stress test, the plasma levels of β-endorphin andPOMC-correlated peptides were not modified. Asan interpretation of this behavior they assumed therise in β-endorphin concentration observed at peakin patients with CAD and negative stress test asassociated with painless ischemia.

Patients with asymptomatic ischemia on exercisehad a significantly greater β-endorphin responsethan those with angina. Public speaking elicited asignificantly larger β-endorphin increase than didexercise. Patients with silent versus painful ischemiaexperience had a greater β-endorphin response toexercise. However, the β-endorphin response to a speech stressor was greater than to exercise.Increased β-endorphin response to a speech stressor

was interpreted as a partial predominance of silentischemia during psychological stress (Miller et al.1993). In patients with CAD and exercise-inducedischemia, public speaking produces psychologicalstress manifested by increased cardiovascular reac-tivity and causes an increase in plasma β-endorphinlevels that was significantly correlated with painthresholds (Sheps et al. 1995). Sex differences inchest pain at exercise conditions were observed by the same authors. Women reported chest painmore often than men during daily activities andduring laboratory mental stressors but not duringexercise. Men had lower scores than women onmeasures of depression or trait anxiety. Women hadsignificantly lower plasma β-endorphin levels atrest and at maximal mental stress. They concludedthat their results reflect sex differences in the affect-ive and discriminative aspects of pain perception,which may help to explain sex-related differences inclinical presentations (Sheps et al. 2001).

Plasma β-endorphin levels were studied due toexercise-induced ischemia in patients with CAD.There was no significant difference in plasma β-endorphin levels during or after exercise betweensymptomatic and asymptomatic patients; thus, thedifferences in circulating levels of β-endorphin andalso ACTH were not associated with the presence orabsence of pain (Heller et al. 1987; Marchant et al.1994). This observation is in accordance to the abovementioned observation of Droste et al. (1991) thatexercise-induced elevation in pain threshold wasnot related to plasma β-endorphin levels.

Influence of age, race and gender

Effects of chronic exertion on β-endorphin and therelationship to melatonin secretion were studied inwell-trained athletes by Appenzeller and Wood(1992): β-endorphin and melatonin increased afterexercise. Interestingly, the magnitude of this in-crease was age-dependent. Chronic exertion wasassociated with a decrease in exercise induced opioid release and in such individuals whose mela-tonin secretion was not β-endorphin related. Thefirst study demonstrating that older men can makephysiological adaptations in the endocrine systemwith resistance training was made by Kraemer, W.J.

Page 160: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

144 chapter 11

et al. (1999). They examined the adaptations of theendocrine system to heavy-resistance training inyounger versus older men in a strength-powertraining program. The amount of cortisol producedat resting levels was reduced and the response to the resistance exercise stress was lower in the oldermen. The decrease in resting concentrations of cor-tisol throughout the training program in the oldermen without significant changes in the ACTH con-centrations indicated that the ACTH receptors in theadrenal gland may have been ‘down-regulated’.Changes in exercise-induced cortisol responsesobserved after exercise were apparently mediatedby a reduction in ACTH responses to exercise stress.The inability to engage similar hormonal mech-anisms in response to heavy-resistance exercise training was interpreted as meaning that the plastic-ity of the endocrine system in older men was alteredor impaired.

Exhausting bicycle exercise in post-menopausalwomen induced a strong release of ACTH, cortisoland prolactin, and natural killer cell activity. Van-der-Pompe et al. (2001) assumed that low vigor in post-menopausal women interferes with theendocrine and immune responses to exhaustingexercise.

Yanovski et al. (2000) have shown that plasmaACTH of African-American women was significantlygreater than that of Caucasian women, but that thiswas not accompanied by greater plasma cortisolconcentrations. Plasma ACTH of African-Americanwomen contained many ‘non-intact’ ACTH frag-ments which were not found in Caucasian women.Plasma ACTH, measured after intense exercise, wassignificantly greater in African-American womenthan in Caucasian women, but plasma cortisol afterexercise was not different. The ACTH of African-American and Caucasian women did not appear tobe equipotent at adrenal melanocortin-2 receptorsbecause the greater ACTH of African-Americanwomen did not lead to greater cortisol secretion.

Data from Goldfarb et al. (1998) suggested thatwomen cycling at 80% Vo2max had a similar β-endorphin response to men independent of theirmenstrual cycle. Comparing ACTH or β-endorphinplasma levels of female athletes with those of maleathletes or volunteers under basal or exercise condi-

tions, no major differences have yet been demon-strated, with the exception of a slightly lower basalβ-endorphin IRM concentration in women as com-pared to men independent of the time of the wom-en’s menstrual cycle.

ACTH, cortisol and β-endorphin were measuredin a treadmill run at 80% of a previously determinedmaximum heart rate of male and female runnersand compared to the concentrations of sedentarymen and women by Kraemer, R.R. et al. (1989). Therun resulted in no rise in β-endorphin, ACTH andcortisol. β-Endorphin values were significantlyhigher in men than in women. No sex or trainingdifferences were seen with respect to change of hor-mone concentrations over the course of the run.Their data indicated that gender and training do notaffect ACTH and cortisol concentrations before,during and after treadmill running at 80% of max-imum heart rate, whereas β-endorphin concentra-tions were higher in men under these conditions.

Plasma ACTH response to exercise was signific-antly attenuated in lactating women performinggraded treadmill exercise finally to elicit 90% Vo2maxuptake in comparison to non-lactating women.These results were interpreted to mean that stress-responsive neurohormonal systems were restrainedin lactating women (Altemus et al. 1995).

HPA and hypothalamic–pituitary–gonadal (HPG)axis modification, combined with cognitive impair-ments, have been reported in elderly subjects andrelated to physical training status. Data from Struderet al. (1999) suggest that elderly endurance athletesreveal a prolonged secretion of glucocorticoids.

Physiological and pathophysiological aspects ofexercise induced POMC release and consequencesto homeostasis of energy and nutrition

Exercise can be regarded as one of many stressorsprovoking metabolic reactions of the organism andtherefore activating the pituitary POMC system.However, in contrast to most of the other stressors,there is no doubt that the organism tries to adapt tothis stressor not by reduction of morphology andfunctions to the original state before exposition toexercise, but by conversion of morphology andfunctions to an altered, i.e. to a stressor-adapted

Page 161: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 145

state, thus counteracting future stress situations ofthe same type.

Sustained hyperglycemia was observed to not be a stimulus to enhanced β-endorphin secretioninto plasma and this lack of response was noteffected by prior exercise (Farrell et al. 1986). Lowerβ-endorphin levels were found in patients with diabetes and silent myocardial ischemia at rest, following exercise, as compared with those withsilent myocardial ischemia who were not diabetic.Hikita et al. (1993) assumed a less significant role of β-endorphin in diabetic patients than in non-diabetic ones.

Angelopoulos (2001) observed comparativelyhigher β-endorphin concentrations during exerciseunder opioid antagonism (naloxone) as comparedto placebo trial and assumed a positive feedbackloop for β-endorphin on plasma glucose regula-tion. Tabata et al. (1991) observed an abolished corticotropin-releasing factor and ACTH increaseafter exercise at workloads of 50% Vo2max untilexhaustion of healthy young men when their bloodglucose concentrations were maintained at the pre-exercise level. The critical level for triggering thepituitary–adrenal–cortical axis was 3.3 mmol. Inderet al. (1998) demonstrated a significant rise in peri-pheral CRH at submaximal exercise, but this wasnot associated with hypoglycemia. Cortisol rise inphysical exercise or during recovery may be initi-ated by the drop in blood glucose and may preventthe inflammatory and immune reactions from over-shooting and so may stabilize homeostasis of theorganism (De Vries et al. 2000).

Numerous studies (for review see Steinberg &Sykes 1985; Cumming & Wheeler 1987; Sforzo 1989;Schwarz & Kindermann 1992; Hoffmann et al. 1996;Goldfarb & Jamurtas 1997; Vassilakopoulos et al.1999) have shown that activation of the pituitaryPOMC system is achieved by a certain degree ofmetabolic demand, which is characterized by bloodlactate levels beyond the anaerobic threshold, which again are reached upon incremental or short-term anaerobic exercise, or which also can bereached after prolonged aerobic endurance exer-cise. Apparently the anaerobic state leads to therelease of CRH and arginin–vasopressin, known tobe released from the hypothalamic paraventricular

nucleus or from the posterior pituitary, respect-ively, for enhancement of POMC fragment release(Inder et al. 1998).

Thus, although the prerequisites or the conse-quences of anaerobic metabolism are apparentlyclosely related to the activation of the POMC system, lactate by itself does not seem to directlystimulate the hypothalamic structures responsiblefor pituitary POMC system activation (Petrides et al.1999). Taylor et al. (1994) assumed base excess as thebest indicator of β-endorphin release or the primarystimulus for activation of the pituitary POMC sys-tem. In contrast, endurance exercise (marathon andsteady-state cycling) reported that constant lactatelevels were not related to β-endorphin increase(Goldfarb et al. 1991; Heitkamp et al. 1996). Bloodlactate levels do not appear to be related to HPAhormone plasma concentrations at high exerciseintensities (Kraemer, W.J. et al. 1989b). It might beassociated with stimulation of testosterone secre-tion or plasma testosterone elevation during exer-cise, but this requires further investigation (Raastadet al. 2000). However, lactate was significantly correlated with β-endorphin, ACTH and cortisol(Kraemer, W.J. et al. 1989a). Low volume resistiveexercise elevates lactate concentrations withoutaltering endorphin (Kraemer, R.R. et al. 1996).

Metabolic response to exercise stress should beenvisaged as a possible candidate for POMC frag-ment function (Weissman 1990). In view of the factthat the release of POMC fragments is obviouslydependent on anaerobic state with lactate levelsbeyond the anaerobic threshold, and that metabolicacidosis probably is a direct stimulus for POMCfragment release, the most likely functional target of POMC fragment release would be counterbalanc-ing the obvious metabolic derailment, exercise isresponsible for, by increased energy supply of theskeletal muscle (Knudtzon 1986; Evans, A.A. et al.1997). Since an anaerobic, acidotic state appears to be the stimulus for β-endorphin release, β-endorphin targets might well be parts of a peri-pheral system responsible for metabolic homeostasisor acid/base equilibrium (Schulz et al. 2000).

Acute amino acid supplementation enhanced the ACTH, luteinizing hormone (LH) and follicle-stimulating hormone (FSH) response to CRH and

Page 162: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

146 chapter 11

gonadotropin-releasing hormone (GnRH) (Di Luigiet al. 1999), whereas acetylsalicylic acid influencesACTH, β-endorphin and cortisol responses to exer-cise-related stress in humans. Their data confirm a role of prostaglandin in these responses. It mightbe of interest, because of the large use of anti-inflammatory drugs in athletes, whether the inter-action between acetylsalicylic acid and hormonesmight positively or negatively influence health status (Di Luigi et al. 2001).

POMC and exercise and immunological influence

Exercise can be regarded as one of many stressorsprovoking immunologic reactions of the organismand therefore activating the pituitary POMC sys-tem. Effects on the immune system elicited by β-endorphin (Teschemacher et al. 1990b) are still amatter of speculation.

An attractive candidate to be considered wouldbe a POMC fragment influence on the immune system. Interactions of β-endorphin (Sibinga &Goldstein 1988) or ACTH (via corticosteroid releasefrom the adrenals) with cells of the immune systemare well testified, such as effects of stress in general(Fricchione & Stefano 1994) and of physical exercisein particular (Jonsdottir et al. 1997). These effectscould contribute to both activation as well as suppression of separate functions of the immunesystem responsible for defense against infections or necessary for morphological alterations in termsof elevated capability to cope with future exercisestress.

It is suggested that exercise can be employed as a model of temporary immune suppression thatoccurs after severe physical stress. The increase incatecholamines and growth hormone mediate theacute effects of exercise on neutrophils, whereascortisol may be responsible for maintaining lym-phopenia and neutrocytosis after exercise of longduration. Lastly, the role of β-endorphin is lessclear, whereas the cytokine response is closelyrelated to muscle damage. However, POMC doesnot seem to be directly involved in the elevatedcytokine level (Pedersen et al. 1997; Pedersen &Hoffmann-Goetz 2000) or elevations of basal naturalkiller cell activity (Dishman et al. 2000). However,

observations from Nagao et al. (2000) suggest thatexercise-induced catecholamines modulate the ex-pression of adhesion molecules on natural killercells, resulting in a mobilization of natural killercells into the circulation.

The mechanisms underlying exercise-associatedimmune changes are multifactorial. Altered plasmaglucose has also been implicated in decreasing stresshormone levels and thereby influencing immunefunction (Nieman & Pedersen 1999).

A further candidate related to stress, immunolo-gical function, insulin sensitivity and cardiovasculardisorders is dehydroepiandrosterone, secreted bythe adrenal cortex in response to ACTH (for reviewsee Kroboth et al. 1999).

Strenuous exercise recruits neutrophils and lym-phocytes into the circulation and, during recovery,lymphocyte concentration rapidly decrease, result-ing in lymphocytopenia if exercise intensity andduration are of sufficient magnitude. A novelfinding of a study by Ronsen et al. (2002) was themore pronounced neutrophilia and lymphocyto-penia during and after a second bout of exercise in the short-rest trial compared to the long-rest trial.They showed that repeating exercise with only afew hours rest results in magnified neuroendocrinestress responses and conceived that performingdaily repeated exercise sessions, thus imposing sub-stantial physical, psychological and metabolic stress,could lead to both adaptive and maladaptive altera-tions in the immune system (Ronsen et al. 2002).

Correlations between POMC derivatives andgonadal steroids or gonadotropins under exercise conditions

To investigate the mechanisms of blunted adrenocor-tical responsiveness to exercise and mild hypercor-tisolism in eumenorrheic and amenorrheic runners in comparison to eumenorrheic sedentary womenACTH stimulation, tests were performed in thepresence and absence of dexamethasone suppres-sion. The cortisol response to stimulation in amen-orrheic runners was blunted in the presence of theirmild hypercortisolism and appeared to be due to a normal limitation in maximal adrenal secretorycapacity (De Souza et al. 1994).

Page 163: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 147

Ovulatory and eumenorrheic runners underwentexercise in the follicular and luteal phases and afterGnRH agonist desensitization. Baseline peripheralβ-endorphin and cortisol levels were not differentbetween the eumenorrheic and oligomenorrheicgroups. A significant increase in β-endorphin levelsin response to exercise occurred only in the eumen-orrheic group after GnRH agonist desensitization.They concluded that alterations in menstrual cyclic-ity and ovulation in conditioned runners were not due to an increase in opioid tone. The hypo-thalamic–gonadotropic axis appeared to be intact inoligomenorrheic runners. Sex steroid administra-tion had no effect on basal β-endorphin levels, butthis probably was not due to pre-existing increasedopioid tone (Meyer et al. 1999).

Menstrual cycle alterations in athletes are accom-panied by shortening of the luteal phase and sec-ondary amenorrhoea. So far it appears that thereduced LH secretion might be caused by an in-creased CRH secretion inhibiting GnRH release. Inaddition, increased CRH tone leads to increased β-endorphin levels which also inhibits GnRH release(Keizer & Rogol 1990).

Effects of oral contraceptives on plasma β-endorphin.Plasma immunoreactive β-endorphin levels at restwere higher in non-pill users than in pill users.Corticotropin levels at rest did not differ betweenthe pill and non-pill users. At 60% Vo2max a slightincrease was found in the concentrations of corti-cotropin and β-endorphin in the non-pill but not inthe pill users. At 90% Vo2max, plasma β-endorphinand corticotropin levels increased significantly inboth groups. A threshold elevation of the intensityof exercise required to increase β-endorphin andcorticotropin secretion by the use of oral contracept-ives was concluded (Rahkila & Laatikainen 1992).To examine individual hormonal responses toextreme physical stress, blood samples were takenfrom highly trained athletes before and withinfinishing a 1000-km ultramarathon foot race. ACTHlevels were significantly elevated above the normalrange. Immunoreactive β-endorphin, growth hor-mone, prolactin, testosterone, cortisol and cortisol-binding globulin were within the normal range.Catecholamines and ACTH remained significantly

elevated above the normal mean and β-endorphinwas within the normal range, without significantchange. A significant increase in cortisol was seen,with no change in cortisol-binding globulin. As amodel of chronic physical stress, the results demon-strated a significantly altered baseline hormonalstate as reflected in the primary mediators of thestress response, the catecholamines and the HPAaxis. The athletes’ response to severe exercise wasdistinct from that of untrained individuals in whoconjugated catecholamines decrease and ACTHincrease. This was interpreted as a possible hor-monal adaptation to prolonged stress (Pestell et al.1989).

New aspects of the functionalsignificance of POMC and interpretationof stress definitions

Functional significance of POMC derivatives

The functional significance of most of the POMCsystems scattered over the organism is unknown. So far this holds for the pituitary POMC system. It has been known for a very long time that thecleavage products of POMC are released into theblood under ‘stress’ conditions. The pituitary glandas an endocrine organ is destined for sending hor-monal messages to tissues of the whole organismvia the cardiovascular compartment, receiving theorders for this kind of message transfer mainly fromthe central nervous system, but also, to a consider-ably smaller extent, over the cardiovascular com-partment itself. Apparently, the pituitary containsseveral message transfer systems thought for specific transfer tasks such as the gonadotropic or the thyreotropic system. For each of these specificmessage transfer systems, specific orders based onspecific stimuli can be postulatedaas well as specifictargets in the periphery to be met by the hormonalmessengers released from the pituitary into theblood (Mooren et al. 2005).

In fact, for the pituitary POMC system a big num-ber of stimuli triggering its activation have beendemonstrated, which can all be comprised underthe term ‘stress’. Physical as well as emotional stres-sors have been shown to induce the release of

Page 164: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

148 chapter 11

POMC fragments into the cardiovascular compart-ment. These stressors range from fear to pain, fromphysical exercise and training to exhaustion exer-cise, from metabolic shifts to serious injury due toparturition, surgery or even accidents. Peptidesderived from POMC are, in function, as hormoneswhen they are secreted from pituitary cells into thebloodstream.

However, it should be mentioned that phys-ical and psychological stressors are processed in the organism in a different way: the informationlaunched upon peripheral tissue injury, in principlecarrying the character of life-threat, is immedi-ately signaled via spinal cord to the brain and isdirectly linked to the pituitary via hypothalamicstructures. In contrast, emotional stressors or thepsychological components of physical stressors arethought to be received and processed by supra-spinal structures such as the limbic system in apretty complicated way before the resulting infor-mation is transferred via hypothalamic structures to the pituitary for activation of the POMC system(Herman & Cullinan 1997), i.e. the activation of theHPA axis.

Since the isolation of the first endogenous opioids in 1975, interest has focused on deducingthe analgesic properties of β-endorphin because the amino acid sequence of its N-terminus is able to bind with opiod receptors. However, exercise-induced elevation in pain threshold was not relatedto plasma endorphin levels (Droste et al. 1991),whereas elevated serum β-endorphin concentra-tions induced by exercise have been linked toaltered pain perception (Harber & Sutton 1984). β-Endorphin has been correlated with several psy-chological and physiological changes, including‘exercise-induced euphoria’, ‘runners’ high’, exer-cise dependence, negative mood state changes, food intake suppression, immune suppression andreproductive dysfunction. But the insulation func-tion provided by the blood brain barrier precludesany association between endorphin and central nervous activity, and, in addition, an increase of β-endorphin associated with an intensive bout ofaerobic exercise appears to be independent of a tendency towards exercise dependence (Pierce et al.1993a).

Methodological aspects of determination ofPOMC derivatives

A large individual variation in the β-endorphin/β-LPH response was noted by Farrell et al. (1982) andSheps et al. (1988). In addition, some methodologicaldifficulties concerning the determination of β-endorphin still exist. Whereas the determination of ACTH always revealed clear-cut results, in many studies β-endorphin IRM, in addition to β-endorphin, was determined to may be contain atleast 10 β-endorphin derivatives. In fact, recentfindings showed that β-endorphin IRM was moreidentical with β-LPH than with β-endorphin. Instudies, wherein β-LPH was determined in com-parison with ACTH or β-endorphin IRM using β-LPH specific determination methods (Oleshansky etal. 1990, Petraglia et al. 1988), β-LPH reached aboutthe same plasma concentrations as β-endorphinIRM or ACTH. In contrast, further studies withselective or even highly specific assays for authenticβ-endorphin showed that under exercise conditionsthe plasma concentrations of authentic β-endorphinwere low (Farrell et al. 1987; Engfred et al. 1994) tominimal (Harbach et al. 2000; Schulz et al. 2000) in comparison with ACTH or β-LPH. Fragmentsfrom different regions of β-endorphin (1–31) mighthave quite different functions, as demonstrated onimmune cells: β-endorphin interacts through its N-terminal fragment with opioid receptors whereasits interactions with binding sites on complement oron thymocytes occur via its C-terminus. Thereforethe identity of ‘β-endorphin’ claimed to occur in theplasma upon exercise is of considerable importancefor correct conclusions in terms of a possible func-tion (Schulz et al. 2000).

Also, POMC fragment release from the adrenalmedulla under physical exercise cannot be excluded(Evans, C.J. et al. 1983). Besides ACTH and β-LPH,further POMC derivatives, which apparently repres-ent small-sized β-endorphin fragments (Wiedemann& Teschemacher 1983), may be released upon phys-ical exercise under certain conditions. The pro-portions of the plasma concentrations of defined β-endorphin fragments may also vary as dependenton the state of fitness of the volunteers (Viru &Tendzegolskis 1995).

Page 165: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 149

In addition to catecholamine release, there is evidence for an acute response of the pituitaryPOMC system to all kinds of stressors. However,there is no clear-cut information about the func-tional significance of the POMC fragments releasedunder stress into the cardiovascular compartment.Although there are well-known targets such as theadrenal gland for ACTH, the functional significanceof the effects elicited by the POMC fragments atperipheral targets is not clear at all; effects by ACTHvia corticosteroids, for example in response to fear,are still a matter of speculation. Thus, the pituitaryPOMC system clearly plays the role of a ‘stress-responder’ but as yet it cannot be classified as a‘stress-adapter’, since it is unclear how it might con-tribute to the reduction of a disturbed function to ahomeostatic state (Teschemacher 2003). Although acouple of well-known exercise effects have beenlinked to POMC fragments, in particular to opioidpeptides (for review see Cumming & Wheeler 1987),the functional significance of the POMC fragmentsreleased under exercise conditions is still a matter of speculation. Some of the functions discussed forPOMC derivatives released into the cardiovascu-lar compartment appear to be unlikely, since therespective effects are not thought to be elicited inperipheral tissues but in the central nervous sys-tem; for example, reduction of depressive state,reduced anxiety, improved self-esteem, improvedwell beingaall in all, an improvement of mood occasionally becoming manifest as ‘runner’s high’.Experimental data indicate that aerobic exercise canactivate central nervous system endogenous opioidsystems, as shown by altered brain opioid levelsand by increased levels of cerebrospinal fluid β-endorphin in running rats (Hoffmann et al. 1996).

The most likely hormonal role for circulating β-endorphin is the modulation of the adrenal responseto stress, by controlling the release of cortisol in re-sponse to ACTH stimulation (McLoughlin et al. 1993).Further candidates for POMC fragment functions inthe periphery are certainly influences on food uptakeor reproduction, since fat tissue or gonads representperipheral targets. However, central POMC sys-tems in hypothalamic areas, in either case, appear tobe more important candidates for influence onreproductive dysfunction (Rivier & Rivest 1991).

β-Endorphin and psychological effects during exercise

No biochemical link was observed for β-endorphinthat might explain the possible influence of physicalactivity on depression (Williams & Getty 1986).However, lower β-endorphin resting plasma levelsof trained subjects were related to an adaptation toexercise training and a greater emotional stabilityand lower depression (Lobstein et al. 1989). Endur-ance training of 8 months duration appeared todecrease the resting plasma β-endorphin concen-trations and the depression scores (Lobstein &Rasmussen 1991).

Singh et al. (1999) observed different plasmaACTH and cortisol responses to psychological andexercise stress tests after dexamethasone treatment.Subjects were classified as responders based onACTH responses to exercise. A psychological stresstest raised heart rate, blood pressure, plasma ACTHand cortisol levels in both high responders (HRs)and low responders (LRs). HRs tended to havehigher heart rates and blood pressures in anticipa-tion of the psychological stress test than LRs. ACTHresponses of HRs were higher, although not sig-nificantly, throughout the psychological stress testthan LRs. HRs had a significantly greater cortisolresponse to the psychological stress than LRs. Theysuggested that the adrenal cortex of the HRs werehypersensitive to ACTH and concluded that menwho are highly responsive to exercise stress werealso highly responsive to psychological stress.

Post-traumatic stress disorder may be associatedwith changes in endogenous opioid peptide func-tion. To test this hypothesis, Vietnam combat veter-ans with post-traumatic stress disorder underwenta standard exercise stress test. Resting plasma β-endorphin levels were comparable between veter-ans and controls. However, post-exercise plasmaβ-endorphin levels were significantly higher thanresting levels only in the post-traumatic stress dis-order patients. These data suggested a differentialalteration in plasma β-endorphin response to exer-cise in post-traumatic stress disorder (Hamner &Hitri 1992). Healthy adolescent women receivedpsychological and endocrine examinations. Anxietyscores and frustration tests were used. On the basis

Page 166: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

150 chapter 11

of the results of these tests, subjects were dividedinto two groups: normal subjects and subjects withevidence of anxiety and/or frustration. Plasma levels of ACTH and β-endorphin were measuredunder basal conditions and after physical exer-cise. Basal concentrations of ACTH and cortisolwere similar in the two groups, whereas basal β-endorphin levels were significantly higher in theanxiety/frustration group than in the control group.A striking increase in plasma ACTH and a slightincrease of β-endorphin levels were observed in theanxiety/frustration group after exercise. Absolutelevels of ACTH and β-endorphin after physicalexercise were significantly higher in this group than in the control group. These findings indicateincreased levels of adrenocorticotropic and opioidactivity in adolescent women with high scores onpsychological measures of anxiety and frustration(Gerra et al. 1992). In addition, significant elevationsof β-endorphin and CRH were observed after running and meditation and were associated as apositive influence (Harte et al. 1995).

However, in one study by McGowan et al. (1993)no significant relationship was observed betweenpre- or post-exercise plasma β-endorphin and eithertotal mood disturbance.

Conclusions

The secretion of POMC or POMC derivatives dur-ing exercise is regarded as an adaptive attempt ofthe athlete’s organism to cope with different stresssituations and is intimately linked to a variety of psy-chological strategies which facilitate its navigationthrough a stressful environment (McCubbin 1993).The secretion of ACTH and β-endorphin and theirincrease occurs in relation to the intensity and theduration of the physical exercise. But the exerciseresponse may also be affected by the training statusof the individual and the population being investig-ated (Goldfarb & Jamurtas 1997).

Efforts to clarify the question of β-endorphin in-volvement in exercise effects by the blockade of opi-oid receptors during exercise (Strassman et al. 1989;Angelopoulos 2001) revealed controversial effectson β-endorphin plasma levels but did not providefurther insight into the mechanisms under question.

In case of situations of overtraining challenge, a reduced ACTH response reflects the athlete’simpaired ability to cope with the stress situation.ACTH and cortisol responses of maximal intensityresistance exercise overtraining are contrary tohigh-volume resistance exercise and overtrainingwith highly aerobic activities. Thus, it becomesapparent that signs of overtraining, based on datafrom endurance athletes, cannot necessarily beapplied to overtraining resulting from highly ana-erobic activities. Future research must address thepossible mechanism(s) of such a cortisol response,whether it is due to adrenal cortex exhaustion orsympathetic or other control (Fry & Kraemer 1997).

POMC fragments may have influence on theimmune system, in particular at physical exercisestress (Jonsdottir et al. 1997). It is suggested thatexercise can be employed as a model of temporaryimmune suppression that occurs after severe phy-sical stress. The increase in catecholamines (epine-phrine and norepinephrine) and growth hormonemediate the acute effects of exercise on neutrophils,whereas cortisol may be responsible for maintain-ing lymphopenia and neutrocytosis after exercise oflong duration (Pedersen et al. 1997). However, therole of β-endorphin is less clear, but the cytokineresponse is closely related to muscle damage, andstress hormones do not seem to be directly involvedin the elevated cytokine level. Lastly, these pheno-mena make it unlikely that β-endorphin plays amajor immune modulatory role in the immediaterecruitment of natural killer cells (for review seePedersen & Hoffman-Goetz 2000).

The metabolic response to exercise stress might bea candidate for POMC fragment function (Weissman1990) in support mechanisms for energy and home-ostasis (Knudtzon 1986). POMC is acting in a com-plex interrelationship between the endocrine,metabolic, cardiac, neurologic and immune systemsunder physical exercise conditions. What can beconcluded from the diverse evidence about exerciseand the involvement of POMC? Given the inherentdifficulties, the following ideas are summarized:1 Among POMC derivatives, the most informationarelated to exercise conditionsais available onACTH, which was studied as the main representa-tive of POMC under the aspect of stress-induced

Page 167: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 151

activation of the HPA axis. Later β-endorphin and β-LPH were recognized to be coreleased from thepituitary into the cardiovascular compartmentunder exercise conditions.2 It is strictly the ‘corticotropic’ part of the pituitaryPOMC system that is activated under physicalstress conditions.3 β-Endorphin plasma levels have universally beenreported to increase with exercise. However, thelarge individual variation in the β-endorphin/β-LPH response and the methodological difficultiesconcerning the determination of authentic β-endor-phin are involved in the problem, and the biologicalsignificance of β-endorphin is still not elucidated.4 Training and extreme physical stress lead to different ACTH and β-endorphin responses. Thestimulus responsible for β-endorphin release underanaerobic exhaustive exercise conditions is prob-ably acidosis, whereas non-exhaustive performance,such as heavy resistance exercise, does not lead toan increase of endorphin levels.5 Age, gender and race have an important influenceon POMC derivatives under exercise conditions,but further investigation of this influence on adapta-tion to acute and chronic exercise is necessary.

6 Glucose and lactate are substrates, which wereintensively observed in exercise physiology becauseof changes under physical and extreme stress.ACTH, cortisol and β-endorphin seem to have aninfluence. However, it is not yet clear in which waythey may contribute in stabilizing the homeostasisof the organism.7 Correlations between POMC derivatives andgonadal steroids or gonadotropins are well testified.8 Exercise is one of many stressors provokingimmunologic reactions and therefore activating thepituitary POMC system. Effects of β-endorphin andACTH can contribute to both activation as well assuppression of the immune system.The response of POMC fragments, such as ACTHand β-endorphin, to exercise stress is complex and,in spite of multiple possible solutions, not yet eluci-dated. Despite evidence for an acute response of the pituitary POMC system to exercise stress (‘stresssensor’), it cannot be classified as a ‘stress regulator’or ‘stress adapter’. Further investigation is neces-saryain addition to the tremendous attempts ofrecent yearsato define the functional significance ofthe POMC fragments released at exercise stress.

References

Adams, H.A. & Hempelmann, G. (1991)The endocrine stress reaction inanesthesia and surgeryaorigin andsignificance. AnaesthesiologieIntensivmedizin NotfallmedizinSchmerztherapie 26(6), 294–305.

Ahtiainen, J.P., Pakarinen, A., Kraemer,W.J. & Häkkinen, K. (2003) Acutehormonal and neuromuscular responsesand recovery to forced vs. maximumrepetitions multiple resistance exercises.International Journal of Sports Medicine24(6), 410–418.

Akil, H., Watson, G., Young, E. et al. (1984)Endogenous opioids: biology andfunction. Annual Revisions of Neuroscience7, 223–255.

Altemus, M., Deuster, P.A., Galliven, E.,Carter, C.S. & Gold, P.W. (1995)Suppression of hypothalamic–pituitary–adrenal axis responses tostress in lactating women. Journal ofClinical Endocrinology & Metabolism80(10), 2954–2959.

Angelopoulos, T.J. (2001) β-Endorphin

immunoreactivity during high-intensityexercise with and without opiateblockade. European Journal of AppliedPhysiology 86(1), 92–96.

Appenzeller, O. & Wood, S.C. (1992)Peptides and exercise at high and low altitudes. International Journal of Sports Medicine 13 (suppl. 1),S135–S140.

Barron, J.L., Noakes, T.D., Levy, W., Smith,C. & Millar, R.P. (1985) Hypothalamicdysfunction in overtrained athletes.Journal of Clinical Endocrinology andMetabolism 60, 803–806.

Bertagna, X. (1994) Proopiomelanocortin-derived peptides. Endocrinology andMetabolism Clinics of North America 23(3),467–485.

Bicknell, A.B., Savva, D. & Luciano, A.A.(1996) Proopiomelanocortin and adrenalfunction. Endocrine Research 22(4),358–393.

Buono, M.J., Yeager, J.E. & Sucec, A.A.(1987) Effect of aerobic training on theplasma ACTH response to exercise.

Journal of Applied Physiology 63(6),2499–2501.

Castro, M.G. & Morrison, E. (1997) Post-translational processing ofproopiomelanocortin in the pituitaryand in the brain. Critical Reviews inNeurobiology 11, 35–57.

Cumming, D.C. & Wheeler, G.D. (1987)Opioids in exercise physiology. Seminarsin Reproductive Endocrinology 5, 171–179.

De Meirleir, K., Naaktgeboren, N., VanSteirteghem, A. et al. (1986) β-Endorphinand ACTH levels in peripheral bloodduring and after aerobic and anaerobicexercise. European Journal of AppliedPhysiology 55(1), 5– 8.

De Souza, M.J., Luciano, A.A., Arce, J.C.,Demers, L.M. & Loucks, A.B. (1994)Clinical tests explain blunted cortisolresponsiveness but not mildhypercortisolism in amenorrheicrunners. Journal of Applied Physiology76(3), 1302–1309.

De Vries, W.R., Bernards, N.T., de Rooij,M.H. & Koppeschaar, H.P. (2000)

Page 168: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

152 chapter 11

Dynamic exercise discloses differenttime-related responses in stresshormones. Psychosomatic Medicine 62(6),866–872.

Denef, C. & Van Bael, A. (1998) A newfamily of growth and differentiationfactors derived from the N-terminaldomain of proopiomelanocortin (N-POMC). Comparative Biochemistry andPhysiology. Part C, Pharmacology,Toxicology & Endocrinology 119, 317–324.

Deschenes, M.R., Giles, J.A., McCoy, R.W.et al. (2002) Neural factors account forstrength decrements observed aftershort-term muscle unloading. AmericanJournal of Physiology. Regulatory,Integrative and Comparative Physiology282(2), R578–R583.

Di Luigi, L., Guidetti, L., Pigozzi F.et al. (1999) Acute amino acidssupplementation enhances pituitaryresponsiveness in athletes. Medicine andScience in Sports and Exercise 31(12),1748–1754.

Di Luigi, L., Guidetti, L., Romanelli, F.,Baldari, C. & Conte, D. (2001)Acetylsalicylic acid inhibits the pituitaryresponse to exercise-related stress inhumans. Medicine and Science in Sportsand Exercise 33(12), 2029–2035.

Diego Acosta, A.M., Garcia, J.C.,Fernandez-Pastor, V.J. et al. (2001)Influence of fitness on the integratedneuroendocrine response to aerobicexercise until exhaustion. Journal ofPhysiology and Biochemistry 57(4), 313–320.

Dishman, R.K., Warren, J.M., Hong, S. et al.(2000) Treadmill exercise training bluntssuppression of splenic natural killer cellcytolysis after footshock. Journal ofApplied Physiology 88(6), 2176–2182.

Donevan, R.H. & Andrew, G.M. (1987)Plasma β-endorphin immunoreactivityduring graded cycle ergometry. Medicineand Science in Sports and Exercise 19(3),229–233.

Droste, C., Greenlee, M.W., Schreck, M. &Roskamm, H. (1991) Experimental painthresholds and plasma β-endorphinlevels during exercise. Medicine andScience in Sports and Exercise 23(3),334–342.

Duclos, M., Corcuff, J.B., Rashedi, M.,Fougere, V. & Manier, G. (1997) Trainedversus untrained men: differentimmediate post-exercise responses ofpituitary adrenal axis. A preliminarystudy. European Journal of AppliedPhysiology 75(4), 343–350.

Duclos, M., Corcuff, J.B., Arsac, L. et al.(1998) Corticotroph axis sensitivity after

Fry, A.C., Kraemer, W.J. & Ramsey, L.T.(1998) Pituitary–adrenal–gonadalresponses to high-intensity resistanceexercise overtraining. Journal of AppliedPhysiology 85(6), 2352–2359.

Ganong, W.F., Dallman, M.F. & Roberts,J.L. (eds.) (1987) Annals of the New YorkAcademy of Sciences, vol. 512. TheHypothalamic–Pituitary–Adrenal AxisRevisited. New York Academy ofSciences, New York.

Gerra, G., Volpi, R., Delsignore, R. et al.(1992) ACTH and β-endorphinresponses to physical exercise inadolescent women tested for anxietyand frustration. Psychiatry Research 41(2),179–186.

Goldfarb, A.H. & Jamurtas, A.Z. (1997) β-Endorphin response to exercise. Anupdate. Sports Medicine 24(1), 8–16.

Goldfarb, A.H., Hatfield, B.D., Potts, J. &Armstrong, D. (1991) β-Endorphin timecourse response to intensity of exercise:effect of training status. InternationalJournal of Sports Medicine 12(3), 264–268.

Goldfarb, A.H., Jamurtas, A.Z., Kamimori,G.H. et al. (1998) Gender effect on β-endorphin response to exercise. Medicineand Science in Sports and Exercise 30(12),1672–1676.

Hamner, M.B. & Hitri, A. (1992) Plasma β-endorphin levels in post-traumaticstress disorder: a preliminary report onresponse to exercise-induced stress.Journal of Neuropsychiatry and ClinicalNeuroscience 4(1), 59–63.

Harbach, H., Hell, K., Gramsch, C. et al.(2000) β-Endorphin (1–31) in the plasmaof male volunteers undergoing physicalexercise. Psychoneuroendocrinology 25(6),551–562.

Harber, V.J. & Sutton, J.R. (1984)Endorphins and exercise. Sports Medicine1(2), 154–171.

Harte, J.L., Eifert, G.H. & Smith, R. (1995)The effects of running and meditation on β-endorphin, corticotropin-releasinghormone and cortisol in plasma, and on mood. Biological Psychology 40(3),251–265.

Heitkamp, H.C., Schmid, K. & Scheib, K. (1993) β-Endorphin andadrenocorticotropic hormoneproduction during marathon andincremental exercise. European Journal of Applied Physiology 66(3), 269–274.

Heitkamp, H.C., Huber, W. & Scheib, K. (1996) β-Endorphin andadrenocorticotrophin after incrementalexercise and marathon runningafemaleresponses. European Journal of AppliedPhysiology 72(5–6), 417–424.

exercise in endurance-trained athletes.Clinical Endocrinology 48(4), 493–501.

Engfred, K., Kjaer, M., Secher, N.H. et al.(1994) Hypoxia and training-inducedadaptation of hormonal responses toexercise in humans. European Journal ofApplied Physiology 68(4), 303–309.

Evans, A.A., Khan, S. & Smith, M.E. (1997)Evidence for a hormonal action of β-endorphin to increase glucose uptake inresting and contracting skeletal muscle.Journal of Endocrinology 155(2), 387–392.

Evans, C.J., Erdelyi, E., Weber, E. &Barchas, J.D. (1983) Identification of pro-opiomelanocortin-derived peptidesin the human adrenal medulla. Science221(4614), 957–960.

Evans, V.R., Manning, A.B., Bernard, L.H.,Chronwall, B.M. & Millington, W.R.(1994) α-Melanocyte-stimulatinghormone and N-acetyl-β-endorphinimmunoreactivities are localized in thehuman pituitary but are not restricted to the zona intermedia. Endocrinology134(1), 97–106.

Facchinetti, F., Panerai, A.E., Petraglia, F. et al. (1987) Proopiomelanocortin-relatedpeptides in feto-placental structuresthroughout pregnancy. ExperimentalClinical Endocrinology 89(1), 48–54.

Farrell, P.A., Gates, W.K., Maksud, M.G. &Morgan, W.P. (1982) Increases in plasma β-endorphin/β-lipotropinimmunoreactivity after treadmillrunning in humans. Journal of AppliedPhysiology 52(5), 1245–1249.

Farrell, P.A., Garthwaite, T.L. & Gustafson,A.B. (1983) Plasma adrenocorticotropinand cortisol responses to submaximaland exhaustive exercise. Journal ofApplied Physiology 55(5), 1441–1444.

Farrell, P.A., Mikines, K.J., Bach, F.W.,Sonne, B. & Galbo, H. (1986) Plasma β-endorphin immunoreactivity: effects of sustained hyperglycemia with andwithout prior exercise. Life Sciences39(11), 965–971.

Farrell, P.A., Kjær, M., Bach, F.W. & Galbo, H. (1987) β-Endorphin andadrenocorticotropin response tosupramaximal treadmill exercise intrained and untrained males. ActaPhysiologica Scandinavica 130(4), 619–625.

Fricchione, G.L. & Stefano, G.B. (1994) The stress response andautoimmunoregulation. AdvancedNeuroimmunology 4(1), 13–27.

Fry, A.C. & Kraemer, W.J. (1997)Resistance exercise overtraining andoverreaching. Neuroendocrineresponses. Sports Medicine 23(2),106–129.

Page 169: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 153

Heitkamp, H.C., Schulz, H., Rocker, K. & Dickhuth, H.H. (1998) Endurancetraining in females: changes in β-endorphin and ACTH. InternationalJournal of Sports Medicine 19(4), 260–264.

Heller, G.V., Garber, C.E., Connolly, M.J. et al. (1987) Plasma β-endorphin levels insilent myocardial ischemia induced byexercise. American Journal of Cardiology59(8), 735–739.

Herman, J.P. & Cullinan, W.E. (1997)Neurocircuitry of stress: central controlof the hypothalamo–pituitary–adrenocortical axis. Trends inNeurosciences 20(2), 78–84.

Hikita, H., Kurita, A., Takase, B. et al.(1993) Usefulness of plasma β-endorphin level, pain threshold andautonomic function in assessing silentmyocardial ischemia in patients withand without diabetes mellitus. AmericanJournal of Cardiology 72(2), 140–143.

Hoffmann, P., Jonsdottir, I.H. & Thoren, P.(1996) Activation of different opioidsystems by muscle activity and exercise.News in Physiological Sciences 11, 223–228.

Höllt, V. (1993) Regulation of opioidpeptide gene expression. In: Handbook ofExperimental Pharmacology (Herz, A.,Akil, H. & Simon, E.J., eds.). Springer,Berlin: 307–346.

Inder, W.J., Hellemans, J., Swanney, M.P.,Prickett, T.C. & Donald, R.A. (1998)Prolonged exercise increases peripheralplasma ACTH, CRH, and AVP in maleathletes. Journal of Applied Physiology85(3), 835–841.

Jonsdottir, I.H., Hoffmann, P. & Thoren, P.(1997) Physical exercise, endogenousopioids and immune function. ActaPhysiologica Scandinavica. Supplementum640, 47–50.

Keizer, H.A. & Rogol, A.D. (1990) Physicalexercise and menstrual cycle alterations.What are the mechanisms? SportsMedicine 10(4), 218–235.

Knudtzon, J. (1986) Effects ofproopiomelanocortin-derived peptideson plasma levels of glucagon, insulinand glucose. Hormonal and MetabolicResearch 18, 579–583.

Koziris, L.P., Kraemer, W.J., Gordon, S.E.,Incledon, T. & Knuttgen, H.G. (2000)Effect of acute postexercise ethanolintoxication on the neuroendocrineresponse to resistance exercise. Journal ofApplied Physiology 88(1), 165–172.

Kraemer, R.R., Blair, S., Kraemer, G.R.& Castracane, V.D. (1989) Effects oftreadmill running on plasma β-endorphin, corticotropin, and cortisollevels in male and female 10 K runners.

suspected and confirmed ischemic heartdisease during exercise. Acta Cardiologica51(1), 27–36.

Lobstein, D.D. & Rasmussen, C.L. (1991) Decreases in resting plasma β-endorphin and depression scores afterendurance training. Journal of SportsMedicine and Physical Fitness 31(4),543–551.

Lobstein, D.D., Rasmussen, C.L., Dunphy,G.E. & Dunphy, M.J. (1989) β-endorphinand components of depression aspowerful discriminators betweenjoggers and sedentary middle-agedmen. Journal of Psychosomatic Research33(3), 293–305.

Loh, Y.P. (1992) Molecular mechanisms ofβ-endorphin biosynthesis. BiochemicalPharmacology 44, 843–849.

Luger, A., Deuster, P.A., Kyle, S.B. et al.(1987) Acute hypothalamic–pituitary–adrenal responses to the stress oftreadmill exercise. Physiologicadaptations to physical training. New England Journal of Medicine316(21), 1309–1315.

Luger, A., Deuster, P.A., Gold, P.W.,Loriaux, D.L. & Chrousos, G.P. (1988)Hormonal responses to the stress ofexercise. Advances in ExperimentalMedicine and Biology 245, 273–280.

McCubbin, J.A. (1993) Stress andendogenous opioids: behavioral andcirculatory interactions. BiologicalPsychology 35, 91–122.

McGowan, R.W., Pierce, E.F., Eastman, N.et al. (1993) β-Endorphins and moodstates during resistance exercise.Perceptual and Motor Skills 76(2), 376–378.

McLoughlin, L., Medback, S. & Grossman,A.B. (1993) Circulating opioids in man. In: Handbook of ExperimentalPharmacology (Herz, A., Akil, H. &Simon, E.J., eds.). Springer, Berlin:673–696.

McMurray, R.G., Forsythe, W.A., Mar,M.H. & Hardy, C.J. (1987) Exerciseintensity-related responses of β-endorphin and catecholamines. Medicineand Science in Sports and Exercise 19(6),570–574.

McMurray, R.G., Fafrowicz, J.F. & Berry,M. (1988) The endorphin response ofwomen to sleep loss and exercise.Aviation, Space, and EnvironmentalMedicine 59(2), 129–132.

Mahler, D.A., Cunningham, L.N., Skrinar,G.S., Kraemer, W.J. & Colice, G.L. (1989)β-endorphin activity and hypercapnicventilatory responsiveness aftermarathon running. Journal of AppliedPhysiology 66(5), 2431–2436.

European Journal of Applied Physiology and Occupational Physiology 58(8),845–851.

Kraemer, R.R., Acevedo, E.O.,Dzewaltowski, D. et al. (1996) Effects of low-volume resistive exercise on β-endorphin and cortisol concentrations.International Journal of Sports Medicine17(1), 12–16.

Kraemer, W.J., Fleck, S.J., Callister, R. et al.(1989a) Training responses of plasma β-endorphin, adrenocorticotropin, andcortisol. Medicine and Science in Sportsand Exercise 21(2), 146–153.

Kraemer, W.J., Patton, J.F., Knuttgen, H.G.et al. (1989b) Hypothalamic–pituitary–adrenal responses to short-durationhigh-intensity cycle exercise. Journal ofApplied Physiology 66(1), 161–166.

Kraemer, W.J., Hamilton, A.J., Gordon,S.E. et al. (1991) Plasma changes in β-endorphin to acute hypobaric hypoxiaand high intensity exercise. Aviation,Space, and Environmental Medicine 62(8),754–758.

Kraemer, W.J., Fry, A.C., Warren, B.J. et al.(1992) Acute hormonal responses in elitejunior weightlifters. International Journalof Sports Medicine 13(2), 103–109.

Kraemer, W.J., Dziados, J.E., Marchitelli,L.J. et al. (1993) Effects of differentheavy-resistance exercise protocols onplasma β-endorphin concentrations.Journal of Applied Physiology 74(1),450–459.

Kraemer, W.J., Häkkinen, K., Newton, R.U.et al. (1999) Effects of heavy-resistancetraining on hormonal response patternsin younger vs. older men. Journal ofApplied Physiology 87(3), 982–992.

Kroboth, P.D., Salek, F.S., Pittenger, A.L.,Fabian, T.J. & Frye, R.F. (1999) DHEAand DHEA-S: a review. Journal of ClinicalPharmacology 39(4), 327–348.

Lac, G., Marquet, P., Chassain, A.P. &Galen, F.X. (1999) Dexamethasone inresting and exercising men. II. Effects on adrenocortical hormones. Journal ofApplied Physiology 87(1), 183–188.

Langenfeld, M.E., Hart, L.S. & Kao, P.C.(1987) Plasma β-endorphin responses to1-hour bicycling and running at 60%Vo2max. Medicine and Science in Sports andExercise 19(2), 83–86.

Lehmann, M., Foster, C., Dickhuth, H.H. & Gastmann, U. (1998) Autonomicimbalance hypothesis and overtrainingsyndrome. Medicine and Science in Sportsand Exercise 30(7), 1140–1145.

Letizia, C., Barilla, F., Cerci, S. et al. (1996)β-Endorphin and propiomelanocortin-correlates peptides response in

Page 170: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

154 chapter 11

Marchant, B., Umachandran, V.,Wilkinson, P. et al. (1994) Reexaminationof the role of endogenous opiates insilent myocardial ischemia. Journal of the American College of Cardiology 23(3),645–651.

Marquet, P., Lac, G., Chassain, A.P.,Habrioux, G. & Galen, F.X. (1999)Dexamethasone in resting andexercising men. I. Effects onbioenergetics, minerals, and relatedhormones. Journal of Applied Physiology87(1), 175–182.

Meyer, W.R., Muoio, D. & Hackney, T.C.(1999) Effect of sex steroids on β-endorphin levels at rest and duringsubmaximal treadmill exercise inanovulatory and ovulatory runners.Fertility and Sterility 71(6), 1085–1091.

el Migdadi, F., Bashir, N., Hasan, Z., alHader, A.A. & Gharaibeh, M. (1996)Exercise at low altitude (Jordan valley)causes changes in serum levels ofACTH, insulin, cortisol and lactate.Endocrinological Research 22(4), 763–767.

Miller, P.F., Light, K.C., Bragdon, E.E. et al.(1993) β-Endorphin response to exerciseand mental stress in patients withischemic heart disease, Journal ofPsychosomatic Research 37(5), 455–465.

Mooren, F., Völker, K., Pedersen, B.K.,Schulz, A. & Teschemacher, H. (2005)Inter- and intracellular signaling. In:Molecular and Cellular Exercise Physiology(Mooren, F.C. & Völker, K., eds). HumanKinetics, Champaign, IL: 109.

Nagao, F., Suzui, M., Takeda, K., Yagita, H.& Okumura, K. (2000) Mobilization ofNK cells by exercise: downmodulationof adhesion molecules on NK cells bycatecholamines. American Journal ofPhysiology. Regulatory, Integrative andComparative Physiology 279(4),R1251–R1256.

Nieman, D.C. & Pedersen, B.K. (1999)Exercise and immune function: recentdevelopment. Sports Medicine 27, 73–80.

Oleshansky, M.A., Zoltick, J.M., Herman,R.H., Mougey, E.H. & Meyerhoff, J.L.(1990) The influence of fitness onneuroendocrine responses to exhaustivetreadmill exercise. European Journal ofApplied Physiology and OccupationalPhysiology 59(6), 405–410.

Owens, P.C. & Smith, R. (1987) Opioidpeptides in blood and cerebrospinalfluid during acute stress. Bailliere’sClinical Endocrinology and Metabolism 1,415–437.

Pedersen, B.K. & Hoffman-Goetz, L. (2000)Exercise and the immune system:

endorphin and corticotropin at rest and during exercise. GynecologicalEndocrinology 6(3), 163–166.

Rahkila, P., Hakala, E., Salminen, K. &Laatikainen, T. (1987) Response ofplasma endorphins to running exercisesin male and female endurance athletes.Medicine and Science in Sports and Exercise19(5), 451–455.

Rahkila, P., Hakala, E., Alen, M., Salminen,K. & Laatikainen, T. (1988) β-Endorphinand corticotropin release is dependenton a threshold intensity of runningexercise in male endurance athletes. LifeSciences 43(6), 551–558.

Rivier, C. & Rivest, S. (1991) Effect of stresson the activity of the hypothalamic–pituitary–gonadal axis: peripheral andcentral mechanisms. Biology andReproduction 45(4), 523–532.

Ronkainen, H.R., Pakarinen, A.J. &Kauppila, A.J. (1986) Adrenocorticalfunction of female endurance runnersand joggers. Medicine and Science inSports and Exercise 18(4), 385–389.

Ronsen, O., Kjeldsen-Kragh, J., Haug, E.,Bahr, R. & Pedersen, B.K. (2002)Recovery time affects immunoendocrineresponses to a second bout of enduranceexercise. American Journal of Physiology.Cell Physiology 283(6), C1612–C1620.

Salzet, M., Salzet-Raveillon, B.,Cocquerelle, C. et al. (1997) Leech immunocytes containproopiomelanocortin: nitric oxide mediates hemolymphproopiomelanocortin processing. Journal of Immunology (Baltimore, Md)159(11), 5400–5411.

Schulz, A., Harbach, H., Katz, N., Geiger,L. & Teschemacher, H. (2000) β-Endorphin immunoreactive materialand authentic β-endorphin in the plasma of males undergoing anaerobicexercise on a rowing ergometer.International Journal of Sports Medicine21(7), 513–517.

Schwarz, L. & Kindermann, W. (1990) β-Endorphin, adrenocorticotropichormone, cortisol and catecholaminesduring aerobic and anaerobic exercise.European Journal of Applied Physiology61(3–4), 165–171.

Schwarz, L. & Kindermann, W. (1992)Changes in β-endorphin levels inresponse to aerobic and anaerobicexercise. Sports Medicine 13(1), 25–36.

Sforzo, G.A. (1989). Opioids and exercise.An update. Sports Medicine 7(2), 109–124.

Shen, A., Chin, J., Fullerton, M., Jennings,G. & Dart, A. (1992) Increases in plasma

regulation, integration, and adaptation.Physiological Reviews 80(3), 1055–1081.

Pedersen, B.K., Bruunsgaard, H., Klokker,M. et al. (1997) Exercise-inducedimmunomodulationapossible roles ofneuroendocrine and metabolic factors.International Journal of Sports Medicine 18(suppl. 1), S2–S7.

Pestell, R.G., Hurley, D.M. & Vandongen,R. (1989) Biochemical and hormonalchanges during a 1000 kmultramarathon. Clinical and ExperimentalPharmacology & Physiology 16(5),353–361.

Petraglia, F., Barletta, C., Facchinetti, F. et al. (1988) Response of circulatingadrenocorticotropin, β-endorphin, β-lipotropin and cortisol to athleticcompetition. Acta Endocrinologica 118(3),332–336.

Petraglia, F., Bacchi, M.A., Comitini, G. et al. (1990) Plasma β-endorphin and β-lipotropin levels increase in welltrained athletes after competition andnon competitive exercise. Journal ofEndocrinological Investigation 13(1),19–23.

Petrides, J.S., Deuster, P.A. & Mueller, G.P.(1999) Lactic acid does not directlyactivate hypothalamic–pituitarycorticotroph function. Proceedings of the Society for Experimental Biology andMedicine 220(2), 100–105.

Pierce, E.F., Eastman, N.W., Tripathi, H.L.,Olson, K.G. & Dewey, W.L. (1993a) β-Endorphin response to enduranceexercise: relationship to exercisedependence. Perceptual and Motor Skills77(3) (Pt. 1), 767–770.

Pierce, E.F., Eastman, N.W., Tripathi, H.T.,Olson, K.G. & Dewey, W.L. (1993b)Plasma β-endorphin immunoreactivity:response to resistance exercise. Journal ofSports Science 11(6), 499–52.

Pierce, E.F., Eastman, N.W., McGowan,R.W. et al. (1994) Resistance exercise decreases β-endorphinimmunoreactivity. British Journal ofSports Medicine 28(3), 164–166.

Raastad, T., Bjoro, T. & Hallen, J. (2000)Hormonal responses to high- andmoderate-intensity strength exercise.European Journal of Applied Physiology82(1–2), 121–128.

Raastad, T., Glomsheller, T., Bjoro, T. &Hallen, J. (2001) Changes in humanskeletal muscle contractility andhormone status during 2 weeks of heavystrength training. European Journal ofApplied Physiology 84(1–2), 54–63.

Rahkila, P. & Laatikainen, T. (1992) Effectof oral contraceptives on plasma β-

Page 171: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

proopiomelanocortin and exercise 155

β-endorphin concentrations duringexercise do not contribute to increases inheart rate following autonomic blockadein man. British Journal of ClinicalPharmacology 33(1), 89–92.

Sheps, D.S., Koch, G., Bragdon, E.E. et al.(1988) The reproducibility of resting andpost exercise plasma β-endorphins. LifeSciences 43(9), 787–791.

Sheps, D.S., Ballenger, M.N., De Gent, G.E.et al. (1995) Psychophysical responses toa speech stressor: correlation of plasmaβ-endorphin levels at rest and afterpsychological stress with thermallymeasured pain threshold in patientswith coronary artery disease. Journal ofthe American College of Cardiology 25(7),1499–1503.

Sheps, D.S., Kaufmann, P.G., Sheffield, D.et al. (2001) Sex differences in chest painin patients with documented coronaryartery disease and exercise-inducedischemia: Results from the PIMI study.American Heart Journal 142(5), 864–871.

Sibinga, A. & Goldstein, A. (1988) Opioidpeptides and opioid receptors in cells ofthe immune system. Annual Reviews ofImmunology 6, 219–249.

Singh, A., Petrides, J.S., Gold, P.W.,Chrousos, G.P. & Deuster, P.A. (1999)Differential hypothalamic–pituitary–adrenal axis reactivity to psychologicaland physical stress. Journal of ClinicalEndocrinology and Metabolism 84(6),1944–1948.

Steinberg, H. & Sykes, E.A. (1985)Introduction to symposium onendorphins and behavioural processes;review of literature on endorphins andexercise. Pharmacology, Biochemistry, andBehavior 23(5), 857–862.

Strassman, R.J., Appenzeller, O., Lewy,A.J., Qualls, C.R. & Peake, G.T. (1989)Increase in plasma melatonin, β-endorphin, and cortisol after a 28.5-milemountain race: relationship toperformance and lack of effect ofnaltrexone. Journal of ClinicalEndocrinology and Metabolism 69(3),540–545.

Struder, H.K., Hollmann, W., Platen, P. et al. (1999) Neuroendocrine system

proliferation following exercise.International Journal of Sports Medicine22(6), 447–453.

Vassilakopoulos, T., Zakynthinos, S. &Roussos, C. (1999) Strenuous resistivebreathing induces proinflammatorycytokines and stimulates the HPA axis inhumans. American Journal of Physiology277(4) (Pt. 2), R1013–R1019.

Viru, A. & Tendzegolskis, Z. (1995) Plasmaendorphin species during dynamicexercise in humans. Clinical Physiology15(1), 73–79.

Walberg-Rankin, J., Franke, W.D. &Gwazdauskas, F.C. (1992) Response of β-endorphin and estradiol to resistanceexercise in females during energybalance and energy restriction.International Journal of Sports Medicine13(7), 542–547.

Weissman, C. (1990) The metabolicresponse to stress: an overview andupdate. Anesthesiology 73(2), 308–327.

Wiedemann, K. & Teschemacher, H. (1983)β-Endorphin immunoreactive materialsin human plasma determined by amultiple radioimmunoassay system. Life Sciences 33 (suppl. 1), 89–92.

Williams, J.M. & Getty, D. (1986) Effect oflevels of exercise on psychological moodstates, physical fitness, and plasma β-endorphin. Perceptual and Motor Skills63(3), 1099–1105.

Wittert, G.A., Livesey, J.H., Espiner, E.A. & Donald, R.A. (1996) Adaptation of the hypothalamopituitary adrenal axisto chronic exercise stress in humans.Medicine and Science in Sports and Exercise28(8), 1015–1019.

Yanovski, J.A., Yanovski, S.Z., Boyle, A.J. et al. (2000) Hypothalamic–pituitary–adrenal axis activity during exercise in African American and Caucasianwomen. Journal of Clinical Endocrinologyand Metabolism 85(8), 2660–2663.

Young, E., Bronstein, D. & Akil, H. (1993)Proopiomelanocortin biosynthesis,processing and secretion: functionalimplications. In: Handbook ofExperimental Pharmacology (Herz, A.,Akil, H. & Simon, E.J., eds.). Springer,Berlin: 393–421.

and mental function in sedentary andendurance-trained elderly males.International Journal of Sports Medicine20(3), 159–166.

Tabata, I., Ogita, F., Miyachi, M. &Shibayama, H. (1991) Effect of low bloodglucose on plasma CRF, ACTH, andcortisol during prolonged physicalexercise. Journal of Applied Physiology71(5), 1807–1812.

Tache, Y. & Rivier, C. (eds.) (1993) Annals of the New York Academy of Sciences, vol.697. Corticotropin-Releasing Factor andCytokines: Role in the Stress Response. New York Academy of Sciences, NewYork.

Taylor, D.V., Boyajian, J.G., James, N. et al.(1994) Acidosis stimulates β-endorphinrelease during exercise. Journal of AppliedPhysiology 77(4), 1913–1918.

Teschemacher, H. (2003)Proopiomelanocortin. What role doesthis ACTH and endorphin precursorplay in stress adaptation? InternistischePraxis 43, 605–615.

Teschemacher, H., Koch, G., Kramer, D. & Wiedemann, K. (1990a) β-Endorphinimmunoreactive material in the plasma:what does it mean? In: Endorphins inReproduction and Stress (Distler, W. &Beck, L., eds.). Springer, Berlin: 93–98.

Teschemacher, H., Koch, G., Scheffler, H.,Hildebrand, A. & Brantl, V. (1990b)Opioid peptides. Immunologicalsignificance? Annals of the New YorkAcademy of Sciences 594, 66–77.

Tharp, G.D. (1975) The role ofglucocorticoids in exercise. Medicine and Science in Sports and Exercise 7(1),6–11.

Urhausen, A., Gabriel, H. & Kindermann,W. (1995) Blood hormones as markers oftraining stress and overtraining. SportsMedicine 20(4), 251–276.

Van-der-Pompe, G., Bernards, N.T.,Kavelaars, A. & Heijnen, C. (2001) Anexploratory study into the effect ofexhausting bicycle exercise on endocrineand immune responses in post-menopausal women: relationshipsbetween vigour and plasma cortisolconcentrations and lymphocyte

Page 172: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

156

Introduction

The insulin-like growth factor (IGF) signaling sys-tem plays a critical role in the growth and develop-ment of many tissues, and is an important mediatorof overall pre- and postnatal growth (reviewed inRosenfeld & Roberts 1999). The IGF system is alsoimplicated in pathophysiology, and plays a particu-larly important role in tumorigenesis. As describedin more detail below, the IGF system is comprised of the IGF ligands (IGF-I and IGF-II), a series of cell-surface receptors that mediate the biologicaleffects of the IGFs, and a family of insulin-likegrowth factor binding protein (IGFBP) that modu-late the half-lives and bioavailability of the IGFs inthe circulation and in extracellular fluids (Fig. 12.1).This review will provide an overview of the IGF

system, its various components and signaling mech-anisms, and its role in growth and development,with an emphasis on human data. As other chaptersin this volume will describe the effects of exercise on specific components of the IGF system, such asthe IGF ligands and the IGFBPs, this chapter willalso discuss the potential effects of exercise on IGFsignaling through the IGF receptors per se.

Insulin-like growth factor systemcomponents

The insulin-like growth factor ligands

igf-i and igf-ii structure

The IGF-I and IGF-II ligands are encoded by large

Chapter 12

Introduction to the Insulin-Like Growth FactorSignaling System

CHARLES T. ROBERTS, JR

16

2 4

35A

L S

A

L S

IGF-I

IGF-II

Insulin

IGF-IRIGFBP-Rs IR IRR IGF-II/M6PR

Fig. 12.1 Schematic overview of theinsulin-like growth factor (IGF)signaling system.

Page 173: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the igf signaling system 157

genes, which have been extensively characterized inhumans and rodents. The mature IGF-I peptide con-sists of B and A domains that are homologous to theB and A chains of insulin. Unlike the situation withinsulin, they are not proteolytically cleaved, butremain linked in the mature peptide by a C domainanalogous to the C peptide of insulin. Both IGF-Iand IGF-II contain an additional short D domainthat is not found in insulin. Additionally, the IGF-Iand IGF-II prohormones contain a C-terminal Epeptide that is cleaved in the Golgi apparatus during secretion. Alternative splicing of exons 5 and6 of the human, rat and mouse IGF-I genes producesalternative E peptides, but the physiological signific-ance of this prohormone diversity is unclear. Thepresence of multiple leader exons in mammalianIGF-I genes also results in variation in the signalpeptide of the preprohormones, but the physiolo-gical consequences of this are also unclear. The organ-ization and splicing of the human and rodent IGF-IIgenes is also complex, but involves non-codingexons and, therefore, does not affect the structure ofthe mature IGF-II molecule or its precursors.

igf-i and igf-ii expression

The expression of the IGF-II gene in rodents iswidespread prenatally, but diminishes drasticallyafter birth, with the choroid plexus and the lepto-meninges being the persistent sites of synthesis inadult animals. Murine expression of IGF-I, on theother hand, is low prenatally and significantlyincreases during puberty, with hepatic productionbeing a major contributor to overall IGF-I levels inthe circulation. IGF-I is produced by numerousother adult organs, however, including kidney,lung and bone, and exerts endocrine, paracrine andautocrine effects. This inverse pattern of IGF-I andIGF-II expression in rats and mice initially led to theconcept of IGF-II as a fetal growth factor and IGF-Ias an adult growth factor. This is not the situation inhumans, however, since both IGF-I and IGF-II areproduced throughout life by multiple tissues. Infact, the circulating levels of IGF-II are consistentlyseveral-fold higher than those of IGF-I, which sup-ports that concept that there are potentially diver-gent roles for the two IGFs in human physiology.

The insulin-like growth factor receptors

The IGF-I and IGF-II ligands interact with an arrayof cell-surface receptors that may be present singlyor in combination on target cells. It was initiallythought that IGF-I primarily activated the type 1IGF or IGF-I receptor (IGF-IR), a transmembranetyrosine kinase structurally and functionally relatedto the insulin receptor (IR) (LeRoith et al. 1995;Adams et al. 2000). IGF-II, on the other hand, wasknown to interact with high affinity with the type 2 IGF, or IGF-II receptor (IGF-IIR). Subsequent studies have shown that both IGF-I and IGF-II inter-act with the IGF-IR, albeit with a ~ threefold differ-ence in affinity (IGF-I > IGF-II). The cloning of theIGF-IIR cDNA revealed that it was identical to thepreviously characterized cation-independent man-nose-6-phosphate (M6P) receptor involved in endo-cytosis and intracellular trafficking of M6P-taggedproteins. Although some early studies proposed an active role for the IGF-IIR in IGF-II signaling,based upon apparent sequence homology betweenthe cytoplasmic domain of the IGF-IIR and the intra-cellular loops of G-protein-coupled receptors, sub-sequent studies ruled out the ability of the shortintracellular domain of the IGF-IIR to mediate sig-nal transduction. The function of this molecule inIGF-II action is thought to reflect its ability to serveas a clearance receptor for the IGF-II, thereby influ-encing the extracellular concentration of IGF-II.

The biological effects of IGF-I result primarilyfrom its activation of the IGF-IR. IGF-I does notcross-react with the IR, except at pharmacologicaldoses, since the relative affinity of IGF-I for the IGF-IR versus the IR differs by at least an order of magni-tude. It was initially thought that IGF-II, like IGF-I,only bound appreciably to the IGF-IR as com-pared to the IR. Studies in knockout mice lackingvarious combinations of the IGF system and the IRsuggested that IGF-II acted through the IR in earlydevelopment, prior to detectable IGF-IR gene ex-pression (Louvi et al. 1997). The molecular basis forthis phenomenon was revealed when it was dis-covered that a splice variant of the IR displayed high affinity for IGF-II. Specifically, the IR transcriptis subject to alternative splicing of exon 11, whichencodes a 12-amino acid segment at the C terminus

Page 174: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

158 chapter 12

of the extracellular β subunit. Previous studies hadshown that the IR isoform encoded by the mRNAlacking the exon-11 sequence (IR-A) displayed atwofold higher affinity for insulin than the IR-B isoform specified by the exon 11-containing mRNA.It has been established that the IR-A isoform, in fact, functions as a high-affinity receptor for IGF-IIand produces predominantly proliferative effects ascompared to the principally metabolic effects elicitedby insulin stimulation of IR-B (Frasca et al. 1999).Thus, IGF-I functions primarily by activating theIGF-IR, while IGF-II can act through either the IGF-IR or through the A form of the IR.

Hybrid receptors and the insulin receptor-related receptor

The scope of IGF signaling is made significantlymore complex by the existence of hybrid receptorsthat result from the dimerization of IGF-IR and IRhemireceptors, each consisting of a single α and βsubunit linked by disulphide bonds (Fig. 12.2).These hybrid receptors are formed by the formation

of intra-α subunit disulphide bonds in the Golgiapparatus of cells expressing both the IGF-IR and IRgenes. While originally considered to represent asmall proportion of the total number of IGF-IR andIR in a given cell, some reports have suggested thatthe formation of hybrids is preferred over the for-mation of classical IGF-IR and IR heterotetramers.This could result from the preferential formation of disulphide bonds between cysteine residues inIGF-IR and IR α subunits themselves. Thus, in somecircumstances, hybrid receptors may outnumber‘pure’ IGF-IR or IR molecules at the cell surface.

With respect to ligand binding, IGF-IR/IR hybridreceptors retain high affinity for IGF-I, but exhibitseverely reduced affinity for insulin. It is thoughtthat this reflects the ability of IGF-I to efficientlybind to either IGF-IR α subunit, while tight insulinbinding requires its interaction with both of the βsubunits found in the IR. As a consequence, the ex-istence of significant number of hybrid receptors may preferentially diminish cellular responsivenessto insulin, but not IGF-I. This has been proposed as amechanism through which up-regulation of IGF-IR

IGF-IR

IGF-IR/IR-A IGF-IR/IR-B IR-A/IR-B IGF-IR/IRR IR-B/IRR

IR-A IR-B IRR

Fig. 12.2 Insulin-like growth factor(IGF) system receptors and hybrids.

Page 175: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the igf signaling system 159

expression could result in insulin resistance in cellsexpressing the IR. The situation with hybrid recep-tors has been further complicated by the existence,and the IGF-II-binding characteristics, of IR-A andIR-B. IR-A/IR-B hybrid receptors undoubtedlyoccur, since most cell express both splice variants.The difficulty in distinguishing these variants experi-mentally has precluded an examination of the bind-ing characteristics and signaling capabilities of thisparticular class of hybrid receptors. It has beendemonstrated, however, that IGF-IR/IR-A hybridsbind IGF-I, IGF-II and insulin, whereas IGF-IR/IR-Bhybrids bind IGF-I with high affinity, IGF-II withlow affinity, and do not bind to insulin (Pandini et al.2002). Thus, the relative expression of the IGF-IRand IR genes and the degree of alternative splicingof exon 11 of the IR gene governs the ability of agiven cell to respond to IGF-I, IGF-II and insulin.

The insulin receptor-related receptor (IRR) is thethird member of the IGF-IR/IR family and does notexhibit binding to IGFs or to insulin (Watt et al.1993). Although still considered an orphan receptor,it has been shown to form hybrids with the IR whenboth entities are overexpressed in NIH-3T3 fibro-blasts. The formal possibility exists, therefore, thatIGF-IR/IRR, IR-A/IRR, or IR-B/IRR hybrids mayoccur in the restricted set of tissues that express theIRR gene, and that the formation of such hybridscould, like the formation of IGF-IR/IR hybrids,influence cellular IGF and insulin responsiveness.In fact, a recent study that analyzed double andtriple knockouts of the IR, IGF-IR and IRR genesdemonstrated a role for the IRR in testis develop-ment, presumably through its modulation of IR andIGF-IR action through hybrid formation (Nef et al.2003).

Insulin-like growth factor binding proteins

The biological activities of the IGF ligands are modulated by a family of high-affinity IGFBPs(IGFBP-1 through -6) that are found in the circula-tion and in extracellular fluids (Jones & Clemmons1995). IGFBP-3 is the predominant IGFBP in serum,and most circulating IGF-I and IGF-II is not found ina free form, but in a ternary complex with IGFBP-3and a third component, the acid-labile subunit

(ALS), in a 1 : 1 : 1 molar ratio. IGFBP-5 also formsternary complexes with IGFs and ALS. While IGFBP-1 through -4 exhibit generally similar affinities forIGF-I and IGF-II, IGFBP-5 and -6 bind IGF-II with10- and 100-fold greater affinities, respectively, thantheir binding to IGF-I. The IGFBPs do not bindinsulin. The IGFBPs control IGF action by increasingthe half-lives of circulating IGFs, by controlling theiravailability for receptor binding, and, in the case of cell surface-associated IGFBPs, by potentiallyinfluencing their direct interaction with receptors.Each of the IGFBPs is subject to limited and poten-tially regulated proteolysis by various proteases.Thus, ligand–receptor interactions in the IGF systemare subject to complex regulation as a result of IGFBPlevels, expression profile, degree of cell–surface asso-ciation and extent of proteolysis.

A series of studies performed over the last several years has established the concept of IGF-independent actions of some of the IGFBPs (Oh1998). IGFBP-3 and 5, in particular, have beenshown to exhibit effects on proliferation, migrationand sensitivity to apoptosis that are independent oftheir effects on IGF signaling per se. Some of these‘IGF-independent’ effects are still modulated by IGF binding to the responsible IGFBP, so that ‘IGFreceptor-independent action’ may be a more accur-ate term for these novel functions. The cell-surfaceor intracellular molecules that participate in thoseeffects are poorly characterized, but IGFBP-3 and -5have been identified in the nuclei of cells followingexposure to exogenous recombinant protein. Thisaspect of IGFBP action, when clarified, will add animportant dimension to our understanding of theIGF signaling system in general.

IGF-I receptor and insulin receptorsignaling pathways

The signaling pathways that mediate IGF action are,in large part, represented by those identified to datefor the IGF-IR (Fig. 12.3). Upon binding of IGF-I or IGF-II to the extracellular α subunit (specifically,a binding region comprised of the internal cysteine-rich region and the adjacent C-terminal L2 domainof the α subunit), a conformational change is inducedin the trans-membrane β subunits, resulting in

Page 176: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

160 chapter 12

trans-autophosphorylation of the tyrosine kinasedomain intrinsic to the cytoplasmic portion of the β subunit. This process fully activates the receptortyrosine kinase, which autophosphorylates the addi-tional tyrosine residues in the juxtamembrane andC-terminal domains that flank the tyrosine kinasedomain. These residues, particularly tyrosine 950 inthe juxtamembrane domain, then serve as dockingsites for members of the insulin receptor substrate(IRS) and the Shc adaptor protein families. Subse-quent phosphorylation of these proteins by thereceptor tyrosine kinase allows IRS and Shc proteinsto engage factors such as Grb-2/SOS and the p85regulatory subunit of PI3 kinase, leading to activa-tion of the MAP kinase and PI3 kinase cascades thatconstitute the major signal transduction cascadesemanating from the activated IGF-IR. Among theultimate targets of the MAP kinase and PI3 kinasecascades are members of the Ets and forkhead tran-scription factor families, which provides a mechan-ism for IGF action at the cell surface to effect thechanges in gene expression that underlie the effectsof IGF signaling on cellular proliferation, differen-tiation and apoptotic sensitivity.

There is a general notion that the MAP kinasepathway exerts effects on proliferation, and that the

PI3 kinase pathway modulates differentiation andsensitivity to apoptosis. However, as is the case withalmost every aspect of the IGF system, nothing issimple. In MCF-7 breast cancer cells, IGF-I-inducedmitogenesis requires the PI3 kinase pathway, butnot the MAP kinase pathway (Dufourny et al. 1997),while in H19-7 neuronal cells, the PI3 kinase path-way is also required for IGF-I-stimulated mitogen-esis, and the MAP kinase cascade is necessary forIGF-I-induced differentiation (Morrione et al. 2000).In terms of anti-apoptotic signaling by the IGF-IR,protection of Rat-1 fibroblasts from ultraviolet-B-induced apoptosis required PI3 kinase pathwayactivation, but not the MAP kinase pathway (Kuliket al. 1997), while IGF-IR-mediated protection ofPC12 cells from serum withdrawal-induced apop-tosis involved both pathways acting in a synergisticfashion (Parrizas et al. 1997). These and other exam-ples suggest that the specific pathways involved in,and their relative contribution to, the effects of IGF-IR signaling on growth, differentiation and apop-tosis are cell type-specific.

While IGF action can clearly be controlled by thelevels of extracellular ligand and the number (andtypes) of receptors at the cell surface, the relativeabundance of receptor targets may be an important

ERK

MEK

Raf

ras

Gab1

Ets AFX

PDK2

PDK1p85

p110

p85

p110IRS-1,2(3,4)

SHC

PKB PKB

Grb-2 sos

Grb-2 sos

Extracellular space

Insulin/IGF-I Rs

Fig. 12.3 Insulin-like growth factor I receptor (IGF-IR) signal transduction pathways.

Page 177: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the igf signaling system 161

factor in determining the effects of IGFs in a giventarget cell. For example, there are four members ofthe IRS family, IRS-1 through 4, each of which has asimilar, yet unique, structure. The presence of dif-ferent combinations of IRS proteins may result indifferential responses to IGF-IR activation. In fact,recent studies have suggested that IRS-3 and IRS-4can inhibit processes activated through IRS-1 andIRS-2 (Tsuruzoe et al. 2001). The relative levels of Shcand IRS proteins may also be an important factorinfluencing IGF action, in that they have beenshown to compete for binding to tyrosine 950 of theactivated IGF-IR.

The general characteristics of signaling and theregulatory possibilities described above for the IGF-IR also apply to the IR (with IR-A and IGF-IR/IRhybrid receptors being relevant to our topic), butthere are important differences between the IGF-IRand IR that may have important ramifications fordifferential actions of IGF-I and IGF-II. In the firstplace, apart from the conserved tyrosine 950/960 inthe IGF-IR and the IR, the position and number of tyrosine residues in the juxtamembrane and C-terminal domains that are subject to autophos-phorylation differ between the IGF-IR and IR. Inaddition, the IGF-IR and the IR have been shown toutilize different heterotrimeric G proteins as part of their signaling mechanisms (Dalle et al. 2001;Kuemmerle & Murthy 2001), and other proteinshave been identified that interact specifically withthe C terminus of the IGF-IR, but not with the IR. Afinal difference in signaling pathways engaged bythe IGF-IR and the IR is the involvement of STAT-3(Zong et al. 1998, 2000; Prisco et al. 2001) and STAT-5(Okajima et al. 1998) in IGF-IR signaling. These dif-ferences, in conjunction with the existence of differ-ent classes of hybrid receptors, make IGF signalingin cells that express the IR (or the IRR), in addition tothe IGF-IR, extremely complicated.

The role of the insulin-like growth factor system in growth anddevelopment

The contributions of IGF action to growth anddevelopment have been discerned from studies intransgenic mice in which various components of theIGF signaling system have been ablated or over-

expressed, and from human studies of populationssuch as Pygmies and rare individuals with muta-tions affecting the IGF-IR and IGF-I genes. Thesefindings are discussed below.

Evidence from transgenic animals

prenatal growth

The role of IGF action in prenatal growth has beeninferred from the phenotypes of transgenic andknockout mice in which the expression of the IGF-I,IGF-II, IGF-IR and IGF-II/MGP receptor genes havebeen manipulated (DeChiara et al. 1991; Baker et al. 1993; Liu et al. 1993; Powell-Braxton et al. 1993).IGF-I and IGF-II deficiency each results in a 40%decrease in birthweight, with IGF-II knockout micealso exhibiting placental growth retardation. Doubleknockouts exhibit an additive growth deficiency of80%. IGF-I knockouts can exhibit general perinatallethality, depending on the genetic background.IGF-IR knockout mice exhibit a 55% decrease ingrowth rate, which is less than that seen in the IGF-I/IGF-II knockouts, and invariably die of suffoca-tion at birth due to inadequate development of themusculature of the diaphragm. Additional deletionof the IGF-I gene in IGF-IR knockout animals doesnot further decrease birth weight, suggesting thatIGF-I functions exclusively through the IGF-IR. Incontrast, IGF-II and IGF-IR double knockouts aremore growth-retarded than single IGF-IR knock-outs, suggesting that IGF-II effects can be mediatedby another receptor during embryogenesis. Ana-lysis of IGF-IR/IR knockouts (Louvi et al. 1997) suggested that the IR was responsible for IGF-II signaling not mediated by the IGF-IR. As mentionedabove, it was subsequently found that alternativesplicing of exon 11 produces an IR isoform thatexhibits high affinity for IGF-II. An indirect role forthe IGF-II/M6P receptor in prenatal growth (Lau et al. 1994; Wang et al. 1994; Ludwig et al. 1997) was inferred from the phenotype of IGF-II/M6Preceptor knockout mice, which exhibit modest fetaland placental overgrowth (25–40%). This pheno-type has been interpreted as resulting from theexcess IGF-II that is seen in the serum and tissues of these animals due to lack of the clearance functionof the IGF-II/M6P receptor.

Page 178: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

162 chapter 12

postnatal growth

IGF-I-deficient mice that do not die perinatallyexhibit severely reduced postnatal growth, whileIGF-II-deficient mice, although smaller than normalat birth, have normal growth velocities postnatally.These findings support the concept that IGF-I is theprincipal mediator of postnatal growth. The lack of a postnatal phenotype in IGF-II-deficient mice isnot surprising in light of the normal shutoff of IGF-IIexpression in almost all mouse tissues postnatally.

Global overexpression of IGF-I in transgenic miceproduces generalized hyperplasia and organome-galy, resulting in adult animals that are 30% largerthan normal (Mathews et al. 1988). Conversely, post-natal overexpression of IGF-II does not result inincreased somatic growth (Rogler et al. 1994; Wolf et al. 1995). Again, the lack of a growth phenotype inIGF-II transgenics may reflect the lack of postnatalIGF-II expression in postnatal rodents.

To date, no convincing phenotype has beenobserved in knockouts of any of the six IGFBPs,including several double knockouts. This puzzlinglack of an effect may reflect the redundancy of func-tion among the six IGFBPs.

Insulin-like growth factor system effects in humans

The role of IGF action in human growth and devel-opment has come from several lines of evidence,including analysis of African Pygmies, a patientwith mutational loss of the IGF-I gene and a series of patients with hemizygosity of the IGF-IR generesulting from loss of the distal arm of chromo-some 15.

efe pygmies

Initial cross-sectional studies of Mbuti and BabingaPygmies concluded that the short stature of thesepopulations was due to the lack of the pubertalgrowth spurt (Mann 1987). Subsequent longitudinalstudies of Efe Pygmies demonstrated growth retar-dation at birth that increased in the first 5 years oflife (Bailey 1990, 1991). More recently, it has beenshown that immortalized T- and B-cell lines fromEfe Pygmies are IGF-I-resistant (Geffner et al. 1993,

1995; Cortez et al. 1996). The molecular basis for this IGF-I resistance and, potentially, the growthphenotype of the Efe population, appears to be adefect in IGF-IR gene expression (Hattori et al. 1996).Thus, decreased IGF action due to decreased IGF-IRlevels causes pre- and postnatal growth retardationin humans.

human mutations affecting the igf-i

and igf-ir genes

One patient has been described who was homozy-gous for a partial deletion of the IGF-I gene (Woodset al. 1996). This patient made no active IGF-I, exhib-ited severe pre- and postnatal growth retardation,and also presented with deafness, mental retarda-tion and microcephaly, characteristics not found inpatients with growth hormone deficiency or resist-ance. This patient’s parents were heterozygous forthe IGF-I gene mutation, had extremely low circu-lating IGF-I levels and also exhibited short stature.These findings provide additional support for a roleof IGF-I in both pre- and postnatal growth anddevelopment.

A number of patients have been described thatare hemizygous for the IGF-IR gene as the result of deletion of the distal arm of chromosome 15(Roback et al. 1991; Siebler et al. 1995) or ring chro-mosome 15 syndrome (Tamura et al. 1993; Peoples et al. 1995). All of these patients exhibited intrauter-ine growth retardation and postnatal growth fail-ure, as well as other developmental abnormalities.Although the growth-deficiency phenotype con-sistently manifested by these patients is consistentwith decreased IGF-IR levels, the fact that no directdemonstration of IGF resistance in cells derivedfrom these patients has been reported makes thisdata supportive, but not definitive, evidence for arole of IGF-IR action in human pre- and postnatalgrowth and development.

Potential effects of exercise on insulin-like growth factor signaling and action

The possible consequences of exercise on the function-ing of the IGF signaling system and IGF-regulatedphysiology can occur at many levels, includingeffects on local and circulating concentrations of IGF

Page 179: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the igf signaling system 163

ligands and IGFBPs (as well as proteolysis of the latter) or more subtle effects on IGF receptor expres-sion and function at a cellular level. The relationshipbetween exercise and the IGFs and their bindingproteins are described in companion chapters, sothe following discussion will focus on exercise and IGF signaling. There are, to date, no data on the relationship between exercise and expression orthe intrinsic activity of the IGF-IR that mediates themajority of IGF signaling.

An alternative possibility is that changes in IRexpression or activity could modulate IGF signal-ing. This could be a direct effect if these changesinvolved the IR-A isoform of the IR, since, as dis-cussed above, this IR isoform is a functional recep-tor for IGF-II. Alteration of expression or activity ofeither IR isoform could also indirectly affect IGF-I orIGF-II signaling through the IGF-IR as a result of theformation of hybrid receptors. While the evidence to date supports an effect of exercise on generalinsulin action and glucose metabolism in particular(reviewed in Wojtaszewski et al. 2002; Zierath 2002),the extent of exercise-induced effects on IR levels or activity, particularly in humans, is unclear. One

study reported an effect of exercise on IR autophos-phorlyation (Youngren et al. 2001), but most studieshave described effects on more distal signaling com-ponents such as IRS-1 and IRS-2 (Chibalin et al. 2000;Nagasaki et al. 2000) and the PI3 kinase pathway (Kimet al. 1998). These latter effects, however, would berather non-specific, as they affect signaling factors util-ized by a multitude of hormones and growth factors.

Conclusions

Despite decades of intensive investigation, thereremain basic aspects of the IGF system that arepoorly understood. Principal among these is therole of IGF-II in human physiology. Additionally,the effects of IGF-I and IGF-II in combination at acellular level are entirely unknown, although mosthuman tissues are routinely exposed to a combina-tion of endocrine, paracrine and, often, autocrineIGF-I and IGF-II. In the context of this volume, thecomplexity of the IGF system provides a wide spec-trum of targets through which the effects of exerciseon IGF-regulated physiology and pathophysiologymay be mediated.

ReferencesAdams, T., Epa, V., Garrett, T. & Ward, C.

(2000) Structure and function of the type1 insulin-like growth factor receptor.Cellular and Molecular Life Sciences: CMLS57, 1050–1093.

Bailey, R.C. (1990) Growth of AfricanPygmies in early childhood. NewEngland Journal of Medicine 323, 1146.

Bailey, R.C. (1991) The comparativegrowth of Efe Pygmies and Africanfarmers from birth to age 5 years. Annalsof Human Biology 18, 113–120.

Baker, J., Liu, J.P., Robertson, E.J. &Efstratiadia, A. (1993) Role of insulin-like growth factors in embryonic andpostnatal growth. Cell 75, 73–82.

Chibalin, A.V., Yu, M., Ryder, J.W. et al.(2000) Exercise-induced changes inexpression and activity of proteinsinvolved in insulin signal transductionin skeletal muscle: differential effects oninsulin-receptor substrates 1 and 2.Proceedings of the National Academy ofSciences of the United States of America 97,38–43.

Cortez, A.B., Van Dop, C., Bailey, R.C. et al. (1996) IGF-I resistance in virus-transformed B-lymphocytes from

African Efe Pygmies. Biochemical andMolecular Medicine 58, 31–36.

Dalle, S., Ricketts, W., Imamura, T.,Vellonweider, P. & Olefsky, J.M. (2001)Insulin and insulin-like growth factor Ireceptors utilize different G proteinsignaling components. Journal ofBiological Chemistry 276, 15 688–15 695.

DeChiara, T.M., Robertson, E.J. &Efstratiadia, A. (1991) Parentalimprinting of the mouse insulin-likegrowth factor II gene. Cell 64, 849–859.

Dufourny, B., Alblas, J., van Teeffelen,H.A.A.M. et al. (1997) Mitogenicsignaling of insulin-like growth factor Iin MCF-7 human breast cancer cellsrequires phosphatidylinositol 3-kinaseand is independent of mitogen-activatedprotein kinase. Journal of BiologicalChemistry 272, 31 163–31 171.

Frasca, F., Pandini, G., Scalia, P. et al. (1999)Insulin receptor isoform A, a newlyrecognized, high-affinity insulin-likegrowth factor II receptor in fetal andcancer cells. Molecular and CellularBiology 19, 3278–3288.

Geffner, M.E., Bailey, R.C., Bersch, N.,Vera, J.C. & Golde, D.W. (1993) Insulin-

like growth factor-I unresponsiveness inan Efe Pygmy. Biochemical and BiophysicalResearch Communications 193, 1216–1223.

Geffner, M.E., Bersch, N., Bailey, R.C. &Golde, D.W. (1995) Insulin-like growthfactor-I resistance in immortalized T-celllines from African Efe Pygmies. Journalof Clinical Endocrinology and Metabolism80, 3732–3738.

Hattori, Y., Vera, J.C., Rivas, C.I. et al.(1996) Decreased insulin-like growthfactor-I (IGF-I) receptor expression andfunction in immortalized African pygmyT-cells. Journal of Clinical Endocrinologyand Metabolism 81, 2257–2263.

Jones, J.I. & Clemmons, D.R. (1995) Insulin-like growth factors and their bindingproteins: biological actions. EndocrineReviews 16, 3–34.

Kim, B., Cheng, H.-L., Margolis, B. &Feldman, E.F. (1998) Insulin receptorsubstrate 2 and Shc play different rolesin insulin-like growth factor I signaling.Journal of Biological Chemistry 273, 34 543–34 550.

Kuemmerle, J.F. & Murthy, K.S. (2001)Coupling of the insulin-like growthfactor-I receptor tyrosine kinase to Gi2 in

Page 180: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

164 chapter 12

human intestinal smooth muscle. Journalof Biological Chemistry 276, 7187–7194.

Kulik, G., Klippel, A. & Weber, M.J. (1997) Antiapoptotic signalling by theinsulin-like growth factor I receptor,phosphatidylinositol 3-kinase, and Akt.Molecular and Cellular Biology 17,1595–1606.

Lau, M.M.H., Stewart, C.E.H., Liu, Z. et al.(1994) Loss of the imprinted IGF2 cation-independent mannnose 6-phosphatereceptor results in fetal overgrowth and perinatal lethality. Genes andDevelopment 8, 2953–2963.

LeRoith, D., Werner, H., Beitner-Johnson,D. & Roberts, C.T., Jr. (1995) Molecularand cellular aspects of the insulin-likegrowth factor I receptor. EndocrineReviews 16, 143–163.

Liu, J., Baker, J., Perkins, A.S., Robertson,E.J. & Efstratiadias, A. (1993) Micecarrying null mutations of the genesencoding insulin-like growth factor I(IGF-1) and type 1 IGF receptor (IGF1R).Cell 75, 59–72.

Louvi, A., Accili, D. & Efstratiadias, A.(1997) Growth-promoting interaction ofIGF-II with the insulin receptor duringmouse embryonic development.Developmental Biology 189, 33–48.

Ludwig, T., Eggenschwiler, J., Fisher, P. et al. (1997) Mouse mutant embryosoverexpressing IGF-II exhibitphenotypic features of the Beckwith–Wiedemann and Simpson–Golabi–Behmel syndromes. Genes andDevelopment 11, 3128–3142.

Mann, G.V. (1987) The riddle of Pygmystature. New England Journal of Medicine317, 709–710.

Mathews, L.S., Hammer, R.E., Behringer,R.R. et al. (1988) Growth enhancement of transgenic mice expressing humaninsulin-like growth factor I.Endocrinology 123, 2827–2833.

Morrione, A., Romano, G., Navarro, M. et al. (2000) Insulin-like growth factor Ireceptor signaling in differentiation ofneuronal H19-7 cells. Cancer Research 60,2263–2272.

Nagasaki, M., Nakai, N., Oshida, Y. et al.(2000) Exercise training preventsmaturation-induced decreases in insulin receptor substrate-1 andphosphatidylinositol 3-kinase in ratskeletal muscle. Metabolism: Clinical andExperimental 49, 954–959.

Nef, S., Verma-Kurvari, S., Merenmies, J. et al. (2003) Testis determination requiresinsulin receptor family function in mice.Nature 426, 291–295.

Oh, Y. (1998) IGF-independent regulation

Tamura, T., Tohma, T., Ohta, T. et al. (1993)Ring chromosome 15 involving deletionof the insulin-like growth factor 1receptor gene in a patient with featuresof Russell–Silver syndrome. ClinicalDysmorphology 2, 106–113.

Tsuruzoe, K., Emkey, R., Kriauciunas,K.M., Ueki, K. & Kahn, C.R. (2001)Insulin receptor substrate 3 (IRS-3) andIRS-4 impair IRS-1- and IRS-2-mediatedsignaling. Molecular and Cellular Biology21, 26–38.

Wang, Z.-Q., Fung, M.R., Barlow, D.P. &Wagner, E.F. (1994) Regulation ofembryonic growth and lysosomaltargeting by the imprinted Igf2/Mprgene. Nature 372, 464–467.

Watt, V.M., Shier, P., Chan, J., Petrisor,B.A. & Mathi, S.K. (1993) IRR: a novelmember of the insulin receptor family.Advances in Experimental Medicine andBiology 343, 125–132.

Wojtaszewski, J.F., Nielsen, J.N. & Richter, E.A. (2002) Invited review:effect of acute exercise on insulinsignaling and action in humans. Journalof Applied Physiology (Bethesda, Md) 93,384–392.

Wolf, E., Rapp, K., Blum, W.F., Kolb, H. & Brem, G. (1995) Skeletal growth oftransgenic mice with elevated levels ofcirculating insulin-like growth factor-II.Growth Regulation 5, 177–183.

Woods, K.A., Camacho-Huebner, C.,Savage, P. & Clark, A.J.L. (1996)Intrauterine growth retardation andpostnatal growth failure associated withthe insulin-like growth factor I gene.New England Journal of Medicine 335,1363–1367.

Youngren, J.F., Keen, S., Kulp, J.L. et al.(2001) Enhanced muscle insulin receptorautophosphorylation with short-termaerobic exercise training. AmericanJournal of Physiology 280, E528–E533.

Zierath, J.R. (2002) Invited review: exercisetraining-induced changes in insulinsignaling in skeletal muscle. Journal ofApplied Physiology (Bethesda, Md) 93,773–781.

Zong, C.S., Zeng, L., Jiang, Y., Sadowski,H. & Wang, L. (1998) Stat3 plays animportant role in oncogenic Ros- andisulin-like growth factor I receptor-induced anchorage-independentgrowth. Journal of Biological Chemistry273, 28 065–28 072.

Zong, C.S., Chan, J., Levy, D.E. et al. (2000)Mechanism of STAT3 activation byinsulin-like growth factor I receptor.Journal of Biological Chemistry 275, 15 099–15 105.

of breast cancer growth by IGF bindingproteins. Breast Cancer Research andTreatment 47, 283–293.

Okajima, Y., Matsumura, I., Nishiura, T. et al. (1998) Insulin-like growth factor-Iaugments erythropoietin-inducedproliferation through enhanced tyrosinephosphorylation of STAT5. Journal ofBiological Chemistry 273, 22 877–22 883.

Pandini, G., Frasca, F., Mineo, R. et al.(2002) Insulin/insulin-like growth factor I hybrid receptors have differentbiological characteristics depending onthe insulin receptor isoform involved.Journal of Biological Chemistry 277, 39 684–39 695.

Parrizas, M., Saltiel, A.R. & LeRoith, D.(1997) Insulin-like growth factor-1inhibits apoptosis using thephosphatidylinositol 3’-kinase andmitogen-activated protein kinasepathways. Journal of Biological Chemistry272, 154–161.

Peoples, R., Milatorich, A. & Francke, U.(1995) Hemizygosity at the insulin-like growth factor I receptor (IGF1R)locus and growth failure in the ringchromosome 15 syndrome. Cytogeneticsand Cell Genetics 70, 228–234.

Powell-Braxton, L., Hollingshead, P.,Warburtin, C. et al. (1993) IGF-I isrequired for normal embryonic growthin mice. Genes and Development 7,2609–2617.

Prisco, M., Peruzzi, F., Belletti, B. &Baserga, R. (2001) Regulation of Id geneexpression by type I insulin-like growthfactor: roles of STAT3 and the tyrosine950 residue of the receptor. Molecular and Cellular Biology 21, 5447–5458.

Roback, E.W., Baraket, A.J., Dev, V.G. et al.(1991) An infant with deletion of thedistal long arm of chromosome 15 (q26.1 -> qter) and loss of insulin-likegrowth factor I receptor gene. AmericanJournal of Medical Genetics 38, 74–79.

Rogler, C.E., Yang, D., Rosetti, L. et al.(1994) Altered body composition andincreased frequency of diversemalignancies in insulin-like growthfactor transgenic mice. Journal ofBiological Chemistry 269, 13 779–13 784.

Rosenfeld, R. & Roberts, C.T., Jr. (1999). TheIGF System. Humana Press, Totowa, NJ.

Siebler, T., Wlodzimierz, L., Terry, C.L. et al. (1995) Insulin-like growth factor Ireceptor expression and function infibroblasts from two patients withdeletion of the distal long arm ofchromosome 15. Journal of ClinicalEndocrinology and Metabolism 80,3447–3457.

Page 181: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

165

Introduction

Physical activity plays an important role in tissueanabolism, growth and development, yet little isknown about the mechanisms that link patterns of exercise with tissue anabolism. Considerableanabolic stimulus arises even from the relativelymodest physical activity of daily living (Leblanc et al. 1990). Therefore, anabolic effects of exercisetraining are not limited to individuals participatingin competitive sports who are particularly focusedon improvement of muscle strength and endurance.On the other hand, participation of young athletesin intense training, especially if associated with in-adequate caloric intake, exposes the young athletesto several health risks and hazards, and may lead toa reduction in growth potential (Theintz et al. 1993).

The exercise-associated anabolic effects are ageand maturity dependent. It is remarkable that nat-urally occurring levels of physical activity, energyexpenditure and muscle strength exhibit some oftheir most rapid increases during childhood andadolescence. The combination of rapid growth and development, high levels of physical activityand spontaneous puberty-related increases in ana-bolic hormones (growth hormone [GH], insulin-likegrowth factor I [IGF-I] and sex steroids) suggest thepossibility of integrated mechanisms linking exer-cise with a variety of anabolic responses. This chap-ter will focus on the effect of physical activity andexercise training on components of the GH–IGF-Iaxis and on differences between systemic and local(i.e. muscle) responses to exercise. Finally, the effectof exercise on the GH–IGF-I axis has been studied

more thoroughly in recent years and, therefore, theGH–IGF-I response to exercise and training canassist competitive athletes and coaches in the evalu-ation of the training load.

The GH–IGF-I axis

The GH–IGF-I axis is composed of hormones,growth factors, binding proteins (BPs) and receptorsthat regulate many essential life processes, includinggrowth and development, metabolic and reparativeprocesses and aging. Therefore, the understandingof the axis must consider each individual com-ponent and the interaction between them underboth physiologic and pathological conditions. Theaxis starts in the central nervous system (CNS)where several neurotransmitters (chatecholamines,serotonin and cholinergic agents, etc.) stimulate the hypothalamus to synthesize growth hormonereleasing hormone (GHRH) and somatostatin (SS).GHRH stimulates the anterior pituitary to synthes-ize and secrete GH. In contrast, SS directly inhibitsGH secretion.

GH is the major secretory product of the axis. Oneof GH most important actions is the stimulation ofhepatic IGF-I synthesis. However, other effects ofGH on metabolism, body composition and tissuedifferentiation are independent of IGF-I. GH exertsdirect feedback effect on the two hypothalamic hor-mones that control its secretion. Tissue GH effectsresult from the interaction between GH and the GHreceptor. The GH receptor contains intracellular andextracellular transmembrane domains. The extra-cellular domain is identical in structure to growth

Chapter 13

Exercise, Training and the GH–IGF-I Axis

ALON ELIAKIM, DAN NEMET AND DAN M. COOPER

Page 182: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

166 chapter 13

hormone binding protein (GHBP) (Leung et al.1987); therefore, a unique feature of this axis is that GH receptor number and activity can be deter-mined easily by measurements of circulating GHBPlevels.

IGF-I is part of the insulin-related peptides. IGF-Ican act as a hormone, and these effects are GHdependent. However, the majority of IGF-I actionsoccur primarily as a result of paracrine or autocrinesecretion and regulation, which are only partiallyGH-dependent. IGF-I is responsible for most, butnot all, of the anabolic and growth promoting effectsof GH. IGF-I stimulates SS secretion, and inhibitsGH by a negative feedback mechanism (Berelowitzet al. 1981), but it is not clear whether circulating orlocal central IGF-I is responsible to this feedbackmechanism.

The majority of circulating IGF-I is bound to several IGFBPs. The most important circulating BPduring adult life is IGFBP-3, which is synthesizedmainly in the liver, and is GH dependent. Whenbound to IGF-I, it complexes with an acid-labile sub-unit to form the circulating complex that carriesmost of the IGF-I in the serum. Some of the IGFBPsare GH dependent (e.g. IGFBP-3), but others, suchas IGFBP-1 and -2, are insulin dependent (beinghigh when insulin levels are low). The interactionbetween IGF-I and its BPs is even more complicatedsince some BPs stimulate (e.g. IGFBP-5), while others inhibit (e.g. IGFBP-4) IGF-I anabolic effects(Rajaram et al. 1997).

The effects of IGF-I result from its interaction with two different receptors. Type I receptor hastyrosine-kinase activity, and mediates most of theIGF-I effects. This receptor exhibit similarities to the insulin receptor and, therefore, may bind alsoinsulin which has known anabolic effects as well.The type II receptor is identical to the mannose-6-phosphate receptor, and binds IGF-II as well.

Some hormones in the GH–IGF-I axis (i.e. GHRH,SS and GH) have a pulsatile pattern of secretion,and it has been shown that the pulsatility of GHsecretion is significantly important for acceleratedgrowth rate (Clark et al. 1985). In contrast, IGF-I andIGFBPs level are relatively stable during the day.

In addition to the important effect on growth, GHand IGF-I have a marked effect on body composi-

tion. Both hormones stimulate increases in musclemass and bone mineral density, and reduce fat distribution.

Several components of the axis are age de-pendent. GHRH, GH, GHBP, IGF-I and IGFBP-3reach their peak circulating levels during puberty(Mauras et al. 1987) and decrease with aging (Corpaset al. 1993). These changes are partially sex-hormonedependent. Nutritional state has also a remarkableinfluence on the GH–IGF-I axis. For example, fast-ing and malnutrition increase GH secretion, butdespite the elevated GH, IGF-I levels are reduced(Marimme et al. 1982), probably due to a decrease in GH receptors. In this chapter we will focus onphysical activity, another environmental regulatorof the GH–IGF-I axis and its components.

The effect of a single exercise

growth hormone

One should differentiate between the acute effectsof a single bout of exercise on the GH–IGF axis fromneuroendocrinological adaptations that occur inphysically active people or in response to long-termprograms of endurance training.

The GH response to exercise is dependent on the duration and intensity of the exercise bout, thefitness level of the exercising subject, the timing of blood sampling, refractoriness of pituitary GHsecretion to exercise stimuli and other environmen-tal factors. Therefore, standardized exercise proto-cols should be used to evaluate the GH response toacute exercise.

Several previous studies reported that the GHresponse to exercise is greater in less fit subjects(Buckler 1972). However, in those earlier studies,subjects were asked to perform exercise tests at the same absolute, rather than relative, power. As aconsequence, due to the great variability in fitness,some subjects exercise below, while others exerciseabove, their lactic/anaerobic threshold (LAT). Thisis important since several investigators (Felsing et al.1992) have demonstrated that circulating GH levelsincreased only in response to above, but not belowLAT, and that exercise loads of 75–90% of maximalaerobic power yielded a greater GH rise than milder

Page 183: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 167

loads (Hartley et al. 1972; Sutton & Lazarus 1976).Therefore, results of studies in which the GHresponse to exercise was tested at an absolute workrate demonstrate simply that as individuals becomefitter, the stress associated with exercise at an abso-lute work rate diminishes.

In contrast to the observation that the exerciseinput should be sufficient to cause a sizeable meta-bolic effect in order to stimulate GH secretion, wepreviously demonstrated a small but significant GHresponse to an exercise input that had no systemiceffect on heart rate or circulating lactate levels (i.e.unilateral wrist flexion) (Eliakim et al. 2000). Thesedata suggest that factors like perceived exertion andassociated psychological stress may lead to activa-tion of the hypothalamic–pituitary axis and GHrelease even in exercises involving small musclegroups.

The duration of exercise for stimulation of GHsecretion should be at least 10 min (Bar-Or 1983),since exercise of shorter duration (e.g. 5 min at aboveLAT [Felsing et al. 1992]) was not accompanied byincreases in circulating GH levels. Moreover, exer-cise-induced GH peak occurs 25–30 min after thestart of the exercise, irrespective of the exerciseduration (Schwarz et al. 1996; Eliakim et al. 1999),and occurs few minutes earlier in women (Widemanet al. 1999). Thus, when the exercise task is brief (e.g.10 min) a peak may be reached after its cessation,while when the exercise task is long (e.g. 45 min) thepeak may be reached while the individual is stillexercising. Blood sampling, however, should betimed to the exercise-induced GH peak.

Pituitary refractoriness, a time in which the normal pituitary gland will not respond sufficientlyto a stimulus for GH release could also influence theGH response to exercise. We previously demon-strated that the GH response to exercise was inhib-ited if a spontaneous, early morning, GH pulse had occurred within 1 h prior to the exercise test(Eliakim et al. 1999). This inhibition occurred prob-ably due to the phenomenon of GH autoinhibition(Pontiroli et al. 1991) in which the elevated circulat-ing GH from the previous spontaneous pulse attenu-ated the pituitary’s response to exercise. Cappon et al. (1994) previously demonstrated a refractoryperiod of at least 1 h following exercise induced GH

secretion (i.e. the subsequent GH response to exer-cise was attenuated), and suggested that exercise-induced elevation in free fatty acids or alterations inparasympathetic–sympathetic tone might have beenresponsible (Casanueva et al. 1984, 1987; Klijman & Frohman 1991). Ronsen et al. (2001) showed arecovery from pituitary refractoriness to GH secre-tion if a second bout of high intensity enduranceexercise was performed 3 h after the first session.Consistent with this report, integrated 1.5-h GHconcentrations were significantly greater if differ-ences between the exercise bouts (30 min, 70%Vo2max) were 3.5 h and not 1 h (Kanaley et al. 1997).

Environmental factors as well as some patholo-gical states may interfere with GH response to exercise. Administration of a high fat meal attenu-ated the magnitude of GH response to exercise(Cappon et al. 1993), and this inhibition was corre-lated with circulating levels of SS. High ambienttemperature may in itself increase circulating GHlevel, while low temperature attenuates GH release(Buckler 1973). Obesity and polycystic ovarian syn-drome (Wilkinson & Parkin 1974) are characterizedby attenuated GH response to exercise. Exercise-associated GH release is reduced in amenorrheicathletes, reflecting possibly decreased GHRH res-ponse to exercise compared to eumenorrheic athletes(Waters et al. 2001). Finally, the age-related declinein GH secretion was associated also with reducedGH response to exercise (Zaccaria et al. 1999).

The increase in circulating GH levels followingexercise may serve in the diagnosis of GH defici-ency. Since GH is secreted in pulses, and duringmost of the day its levels are very low, a single ran-dom blood sample can not differentiate between thehealthy and GH-deficient child. To overcome this, anumber of provocative tests to stimulate pituitaryGH release have been developed (Fraiser 1974).Most of these provocative tests use pharmacologicalagents (Cowell 1995) and present some risk (e.g.hypoglycemia) for the patient. Moreover, the inter-pretation of a normal GH response to pharmacolo-gical stimuli can be questioned because it does notnecessarily give information about physiological GHsecretion. These confounding factors have led anumber of investigators to emphasize the role ofphysiological stimulation tests such as exercise, or

Page 184: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

168 chapter 13

to focus on less variable circulating substances, like IGF-I and/or its BPs, in the diagnosis of GH-deficiency in children (Rosenfeld et al. 1995).

insulin-like growth factors

It is now well established that acute exercise lead toan increase in circulating IGF-I levels. Interestingly,several studies demonstrated that the exercise-induced IGF-I increase occurred following veryshort high intensity exercise (i.e. 90 s) (Kraemer et al.2000), occurred 10 min following the beginning ofendurance exercise (Bang et al. 1990; Schwarz et al.1996) and occurred in exercise of both below andabove the LAT (Schwarz et al. 1996). Exercise wasalso associated with an increase in urinary IGF-I (DePalo et al. 2002).

The mechanism for the transient increase in cir-culating IGF-I in response to exercise is not read-ily apparent. One possibility would be the classicmechanism of increased hepatic IGF release due toexercise-induced secretion of GH. As noted earlier,GH increases significantly mainly in response tohigh intensity exercise, while IGF-I increases forboth low and high intensity exercise. Moreover, circulating IGF-I reaches its peak before the GH peak(i.e. 10 vs. 30 min) (Fig. 13.1), while increases inserum IGF-I, due to de novo IGF-I synthesis in theliver and transport to the circulation, occurs sev-eral hours after the administration of endogenous

GH (Marcus et al. 1990). In addition, earlier stud-ies showed (Bang et al. 1990) that exercise led toincreases in IGF even in subjects with pituitaryinsufficiency. These studies suggest that the exer-cise-associated increase in IGF-I is, in fact, notrelated to GH and must reflect rapid changes in IGF-I distribution in the circulation due to releasefrom marginal pools or changes in IGF-I removal. In addition, the transient nature of the increasessuggests that hemodynamic or metabolic effects ofexercise might play a role. Exercise in humans is accompanied by the rapid autotransfusion ofhemoconcentrated blood from the spleen into thecellular circulation (Flamm et al. 1990), by increasedblood flow to the exercising muscle and by loss ofplasma water (Convertino et al. 1981). Each of thesephenomenona might explain, in part, an increasedIGF concentration by changes in IGF flux and/or volume of distribution.

Another possible source for the increase in circu-lating IGF-I can be a release from the exercisingmuscle. To test this, we used a simple approach inwhich subjects performed a unilateral repeatedflexion of the wrist against relatively high resist-ance, while during- and post-exercise blood sampleswere collected simultaneously from the basilic veinof both the exercising (representing local release)and resting arm (representing systemic response)(Eliakim et al. 2000). We found a bilateral, simul-taneous increase in IGF-I suggesting that the local

–10 00

10 20Time (min)

Exercise

30 40 50 60 70

14

12

10

8

6

4

2

0

340

320

300

280

260

240

220

GH

(n

g·m

L–1)

IGF-

I (n

g·m

L–1)

GH

IGF-I

Fig. 13.1 Typical growth hormone(GH) and insulin-like growth factorI (IGF-I) response to intenseendurance-type exercise bout. PeakIGF-I level occurs before the GH peak emphasizing that the exercise-induced increase in circulating IGF-Iis GH-independent.

Page 185: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 169

exercising muscle was not the source for of the IGF-Iincrease.

Interestingly, Elias et al. (2000) showed a transientrise in circulating IGF-I immediately after exercisetest to exhaustion. IGF-I levels fell thereafter toreach nadir level 60–90 min following the exercise,and then returned gradually to baseline levels.Consistent with this observation prolonged andintense exercise sessions (1.5 h of intense soccerpractice in children [Scheet et al. 1999] or 1.5 h ofwrestling practice in adolescents [Nemet et al. 2002])were associated with decreases in circulating IGF-Ilevels. Interestingly, these sessions were associatedwith increases in proinflammatory cytokines, andthe authors suggested that the increase in theseproinflammatory cytokines mediated the decreasein circulating IGF-I.

The effect of resistance exercise on circulatingIGF-I is inconsistent. Several studies found anincrease in circulating IGF-I and free IGF-I follow-ing strength exercise (Bermon et al. 1999). Moreover,eccentric exercise was associated with increases inmuscle IGF-I mRNA suggesting that IGF-I maymodulate tissue regeneration after mechanical dam-age (Bamman et al. 2001). However, other studiesfound no change in circulating IGF-I followingheavy resistance exercise (Nindl et al. 2001), or evenreduced IGF-I levels the morning after high andmoderate intensity resistance workout (Raastad et al. 2000). These conflicting results reflect probablydifferences in exercise protocols, fitness level of theparticipants, timing of blood sampling, etc.

There have been far fewer investigations inhumans regarding the physiological responses ofIGF-II compared with IGF-I. Several studies (Bang et al. 1990; Schwartz et al. 1996) showed an acute,endurance exercise associated rise in IGF-II. Cir-culating IGF-II may play an important role in bonegrowth and development (Mohan & Baylink 1991);however, the biological importance of the exercise-induced increase in IGF-II has yet to be determined.

insulin-like growth factor

binding proteins

Several studies found increases in IGFBP-3 follow-ing endurance exercise (Schwarz et al. 1996; Chadanet al. 1999). Interestingly, Schwarz and coworkers

have demonstrated that IGFBP-3 levels measuredby radioimmunoassay (RIA) increased with bothlow and high intensity exercise, and were greaterduring the above-LAT protocols (Schwarz et al. 1996).In contrast to their RIA measurements, IGFBP-3measured by Western ligand blotting (WLB) did not change. As a potential explanation for the dis-crepancy between RIA and WLB data they sug-gested that the antibody used in the RIA recognizesboth intact and fragment forms of IGFBP-3 while the WLB method measures only the intact form of IGFBP-3. They therefore measured the rate ofIGFBP-3 proteolysis as a function of exercise andwere intrigued to find that proteolysis did occur inthe high intensity exercise, and that this increasedproteolysis was associated with the peak increase inIGF-I and IGF-II serum concentrations. It is import-ant, however, to note that increases in IGFBP-3 pro-teolytic activity occurred in this study only for highintensity exercise, while increases in IGFBP-3, IGF-Iand IGF-II occurred for both low and high intensityexercise. Moreover, other studies did not find sig-nificant increase in IGF-I proteolytic activity follow-ing endurance exercise (Dall et al. 2001).

The mechanism for the increased IGFBP-3 proteo-lysis following high intensity exercise is not clearlyunderstood. It was suggested that changes in totaland free-ionized serum calcium concentrations(Lamson et al. 1993) and marked serum and muscleacid-base changes (Martin & Baxter 1986) play a rolein exercise-induced IGFBP-3 proteolysis.

Few studies demonstrated changes in otherIGFBPs following exercise, including increases inIGFBP-1 (Suikkary et al. 1989; Hopkins et al. 1994)and IGFBP-2 (Chadan et al. 1999; Nindl et al. 2001).These BPs exist in the circulation in much smallerquantities than IGFBP-3, and appear to play a lesserrole in circulating IGF bioavailability. However, it isclear that the exercise-induced effect on circulatingIGF-I is not only mediated by alteration of theamount of IGF-I but rather by the effect on its BPs.

The effect of exercise training on the GH–IGF-I axis

Several independent studies of healthy human beingshave demonstrated significant correlation betweenphysical fitness and circulating components of the

Page 186: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

170 chapter 13

GH–IGF-I axis. Weltman et al. (1994) demonstratedsignificant positive correlation between 24-h integ-rated GH concentrations and peak Vo2 in younghealthy male, but not female, adults. They alsodemonstrated inverse correlation between 24-hintegrated GH concentrations and body fat in bothmen and women. They suggested that hyper-insulinemia associated with excess body fat anddecreases levels of physical activity may reduce GH release (Yamashita & Melmed 1986), and sincemen, in general, have more central fat than women(Bouchard & Despres 1989), there is a greaterinfluence of adiposity on GH secretion in men. Theyfurther speculated that the higher estradiol levels infemale adults, which stimulate GH release (Ho et al.1987), may oppose the inhibitory effect of insulinand therefore explain the gender-related differencesof the correlation between fitness and GH con-centration. Consistent with this hypothesis, werecently demonstrated in healthy prepubertal girls a remarkable relationship between adiposity (deter-mined by the body mass index [BMI] percentile),fitness and indirect indicators of GH responsiveness(i.e. GHBP) and insulin sensitivity (i.e. IGFBP-1)(Eliakim et al. 2001). It appears that above about the 70 percentile of BMI for age, fitness, GH levelsand insulin sensitivity all begin to decrease even in healthy children (Fig. 13.2). Such informationmight prove to be clinically useful in identifyingchildren who would benefit from programs of phys-ical activity.

The positive correlation between fitness and GHlevels is consistent with animal experiments ofBorer et al. (1986) who noted increased GH pulseamplitude in physically active, rapidly growinghamsters compared with sedentary controls. Theysuggested that the fit state was associated withincreased circulating endorphins and/or increasedtissue sensitivity to endorphins. Endorphins inhibitSS and reduce the inhibition of pituitary GH secre-tion, and as a consequence pituitary GH secretion isincreased.

We previously described that both functional (i.e.Vo2max) and structural (i.e. thigh muscle volumedetermined by magnetic resonance images) indicesof fitness were correlated with mean overnight GHlevels, GHBP and serum IGF-I levels in pre- and

late-pubertal girls (Eliakim et al. 1996, 2001). Thesecross-sectional data suggest that fitness in healthy,prepubertal and adolescent girls is associated withanabolic adaptations of the GH–IGF-I system.

The significant correlation between fitness andmean overnight GH levels resulted probably froman increase in peak GH amplitude since only peakamplitude (and not peak frequency or width) corre-lated with mean GH.

The positive correlation between GHBP and fitnessis unique. GHBP is the extracellular domain of theGH receptor (Rosenfeld 1994), and therefore reflect

0 20 40 60 80 100

160

120

80

40

0

IGFB

P-1

(ng

·mL–1

)0 20 40 60 80 100

800

600

400

200

0

GH

BP

(ng

·mL–1

)

0 20 40 60BMI (percentile)

80 100

60

40

20

0V· O2·

kg−1

(m

L·m

in−1

·kg

–1)

Fig. 13.2 Relationship between age-adjusted body massindex (BMI) percentiles and insulin-like growth factorbinding protein-1 (IGFBP-1) (top panel), growth hormonebinding protein (GHBP) (middle panel), and Vo2peak·kg–1

(bottom panel). The data suggest that there exists a BMI percentile threshold (~ 70%) above which insulinsensitivity, GH activity and fitness all begin to decrease inotherwise healthy, prepubertal girls.

Page 187: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 171

tissue GH receptor capacity. Ligand-mediatedreceptor regulation appears to exist for GH andGHBP in a number of situations. GHBP decreases inacromegaly (Kratzsch et al. 1995) and during exogen-ous recombinant human growth hormone (rhGH)therapy (Legar et al. 1995), and is high in obesitydespite low GH (Jorgensen et al. 1995). But ligand-mediated receptor down-regulation does not ap-pear to operate during normal growth when bothGH and GHBP increase in the prepubertal years(Baumman et al. 1989). Our data suggest that the relatively trained or fit state in prepubertal and adolescent girls is another example of simultaneousincreases in both GH and GHBP. The mechanism ofthese responses is not known, but suggests anabolicadaptations of both the ligand and receptor.

Collectively, it seems that increasing levels ofphysical activity stimulate GH pulsatility and, as a consequence, circulating IGF-I. It is compelling to speculate that the stimulation of the GH–IGF-Iaxis by exercise contributesaalong with genetic,nutritional and other environmental factorsato anincrease in muscle mass and, ultimately, to improvedcardiorespiratory responses to exercise (such asVo2peak). Our data suggest that this mechanismoperate in prepubertal and adolescent girls even asspontaneous growth proceeds.

Significant positive correlation between Vo2maxand both circulating GH and IGF-I levels were alsofound in healthy pre- and post-menopausal women(Kelley et al. 1990). Both Vo2max and IGF-I concen-trations decline with age. However, when theinfluence of both age and fitness was analyzed usingmultiple regression, Vo2max remained the only inde-pendent predictor of circulating IGF-I. Therefore, itwas concluded that the decrease in serum IGF-I wasprobably the result of age related decline in physicalactivity and fitness, and was not related to aging per se. Along with these observations, positive cor-relation between Vo2max and circulating IGF-I werereported in young and old male adults (Poehlman &Copeland 1990), and higher levels of IGF-I werefound in trained middle-aged men (Manetta et al.2002).

Virtually all of the major IGFBPs (1–6), each ofwhich is known to influence IGF-I bioactivity in dif-ferent ways, were also related to indexes of fitness,

suggesting that the BPs may play a role in the inter-action between growth and exercise. With theexception of IGFBP-3, the amount of IGFBPs in thecirculation is low, and the exact role of these IGFBPsin the circulation has yet to be determined. None-theless, it is noteworthy that the cross-sectional relationships between fitness and IGFBPs were con-sistent with current understanding of the biologicalactivity of IGF-I binding proteins based on tissuestudies. IGFBP-1 and -2, known to inhibit IGF-Ifunction, were found to be inversely correlated withmuscle mass (Eliakim et al. 1996, 2001). IGFBP-4, aknown inhibitor of the anabolic functions of IGF-I inbone tissue culture experiments (Mohan et al. 1995),was found to be inversely correlated with musclemass (Eliakim et al. 1996) and Vo2max (Eliakim et al.1998a). In contrast, the IGF-I potentiating bindingprotein, IGFBP-5, was positively correlated withmuscle mass (Eliakim et al. 2001). Accordingly, thesedata suggest the possibility that the generallyincreased IGF-I bioactivity in fitter subjects mightnot be related only to changes in circulating IGF-Ibut also to changes in IGFBPs.

Very few studies examined the effect of endur-ance training on the GH–IGF-I axis longitudinally.Smith, A.T. et al. (1987) studied a group of healthyyoung adult men for 10 days and found thatincreased physical activity exacerbated the well-described reduction in IGF-I that accompaniescaloric restriction. Reduced IGF-I associated withtraining has been observed in high school wrestlersand in highly trained young female gymnasts(Jahreis et al. 1991; Roemmich & Sinning 1997). Inthese studies the training program was accom-panied by loss of body mass providing clear evid-ence for a negative energy balance and catabolicstate. However, a recent report demonstrated thatwhile inadequate caloric intake and negative energybalance is a major cause for the training-associatedIGF-I decrease, IGF-I level may fall even whenenergy balance and weight stability are maintained(Nemet et al. 2004).

We recently reported the effect of a brief (5 weeks)randomized, prospective endurance-type trainingintervention on the GH–IGF-I axis in pre- and late-pubertal boys and girls. Based on the cross-sectionaldata, we hypothesized that training would lead

Page 188: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

172 chapter 13

to increases in circulating GH and IGF-I levels.Training was accompanied by about 15% highertotal energy expenditure (by the doubly labeledwater technique), and resulted in significant increasesin Vo2max and thigh muscle volume in the trainedbut not the control subjects (Eliakim et al. 1996, 1998,2001; Scheet et al. 2002). In contrast to our hypo-theses, there was a significant decrease in GHBP,IGF-I and IGFBP-3 in the trained prepubertal girls(Eliakim et al. 2001); a significant decrease in IGF-Iand IGFBP-3, with a significant increase in IGFBP-2in the trained prepubertal boys (Scheet et al. 2002); a significant decrease in IGF-I and IGFBP-5 in thetrained late-pubertal girls (Eliakim et al. 1996); and a significant decrease in GHBP and IGF-I, with asignificant increase in IGFBP-2 in the trained late-pubertal boys (Fig. 13.3) (Eliakim et al. 1998b).Interestingly, endurance training had no effect onGH pulsatility patterns in any of these groups.

These effects are commonly observed in energy-deficient states like food deprivation or disease-associated malnutrition (Smith, W.J. et al. 1995;Tonshoff et al. 1995), but ‘catabolic’ neuroendocrineadjustments occurred in the present studies eventhough training increased thigh muscle volume.Moreover, despite the significantly greater energy

expenditure in the training group, the exercise inter-vention was not accompanied by weight loss.

A potential mechanism for this seemingly un-expected systemic ‘catabolic-type’ adaptation wasthe reduction in circulating GHBP in the trainedsubjects. As noted earlier, lower GHBP circulatinglevels may reflect fewer tissue receptors and reducedtissue responsiveness to GH (Rosenfeld 1994). As a consequence, since circulating IGF-I depend onGH-induced hepatic production (Lowe 1991), IGF-Ilevels in the training group decreased.

The exercise training effect on GHBP observedhere appears to be unique. In several pathologicalstates (malnutrition [Postel-Vinay et al. 1995], obes-ity [Jorgensen et al. 1995]) the relationship betweenGH and GHBP suggest the well-described phe-nomenon of ligand-induced receptor regulation. Instudies (Eliakim et al. 1998b, 2001), training did leadto a reduction in GHBP, but without any change inmean GH or GH pulsatility. Thus, the possibilityexists that lower GH receptor is the initiating mech-anism in the growth axis response to training. Themechanism for such a direct, training effect on GHreceptors or GHBPs is not known.

Interestingly, training was associated with in-creases in IGFBP-2, a known inhibitor of IGF-I ac-tion, and with reduction in IGFBP-5 in adolescentwomen. IGFBP-5 is one of the binding proteins that enhance some of the IGF-I mitogenic effects (i.e.in bones [Mohan et al. 1995]). These observationsemphasize again that the exercise training induceddecrease in IGF-I bioavailability is mediated notonly by acting on IGF-I itself but by altering its binding proteins.

Recently, Scheet et al. (2002) intriguingly suggestedthe hypothesis that proinflammatory cytokineswere involved in the training-induced decreases of IGF-I (Fig. 13.4) (Scheet et al. 2002). They demon-strated that in brief 5-week endurance training inprepubertal boys increases in Vo2max were pos-itively correlated with changes in tumor necrosisfactor-α (TNF-α). This suggested that children whotrained the hardest and had the biggest increase infitness also had the largest increase in circulatinglevel of proinflammatory cytokines. In addition,changes in IGFBP-3 were inversely correlated withchanges in TNF-α and interleukin-6 (IL-6), suggesting

Prepubertalgirls

Prepubertalboys

AdolescentboysAdolescent

girls

Trained

Control*

* * *

30

25

20

15

10

5

0

–5

–10

–15

–20

–25

IGF-

I (%

ch

ang

e)

Fig. 13.3 Effect of a brief endurance-type exercise trainingon circulating insulin-like growth factor I (IGF-I) levels inprepubertal and adolescent boys and girls. Training wasassociated with a significant decrease or attenuated IGF-Iresponse in all groups.

Page 189: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 173

that increase in the inflammatory response mediatesthe training-associated decrease of components ofthe GH–IGF-I axis.

These observations suggest the hypothesis that a sudden imposition of a training program which is associated with substantial increase in energyexpenditure leads initially to an increase in pro-inflammatory cytokines and, as a consequence, todecreases in IGF-I levels. Further, if the trainingadaptation is successful, the proinflammatory cyto-kines fall, and with that decrease, the suppression of IGF-I diminishes, an anabolic ‘rebound’ in theGH–IGF-I axis may occur, and IGF-I level exceedthe pretraining level. Exactly how and when thisswitch takes place, and whether the initial catabo-lic-type stage is necessary for the ultimate anabolicadaptation, remains unknown.

Adaptations to the training-induced increase inproinflammatory cytokines and decrease in circu-lating IGF-I start early with a training-associatedincrease in IGF-I receptor binding capacity (Lee et al.2000). This increase in IGF-I affinity probably reflectsthe phenomenon of ligand-mediated receptor up-regulation.

Fitter subjects were found to have lower levels of IL-1 receptor antagonist (IL-1ra). This agent isstimulated by the inflammatory cytokines and act toblock their biological activity at the receptor level.Therefore, training-associated reduced inflammat-

ory response leads to lower level cytokines and to alower steady state level of IL-1ra. Finally, and con-sistent with the two phases hypothesis, longer peri-ods of training (5 months [Koziris et al. 1999] and 1 year [Weltman et al. 1992]) were indeed associatedwith increases in circulating GH and IGF-I levels.

Very few studies have examined the effect ofresistance training on the GH–IGF-I axis. Raastad et al. (2001) demonstrated that despite an increase in muscle strength following 2 weeks of heavystrength training, IGF-I decreased in the 8th day andreturned to baseline levels 4 days later. Kraemer et al. (1999) found no change in circulating IGF-I butan increase in resting IGFBP-3 following 10 weeks ofresistance training program in young men. Twelveweeks of high-volume resistance training resultedin type I and type II muscle fiber hypertrophy in college men, and there was a significant correla-tion between the muscle hypertrophy and training-associated increase in GH level (McCall et al. 1999).Longer resistance training programs (25 weeks)were associated with increase in serum IGF-I afterthe 13th week of training in young adults (Borst et al.2001). Since increase in muscle strength occurredalso mainly during the first 13 weeks, the authorssuggested that IGF-I mediated, at least partially, theresistance training-associated improvement in mus-cle strength. These results suggest that a biphasicchange in circulating IGF-I occurs also in responseto resistance training in young adults. In contrast,no changes in circulating IGF-I was found in oldadults (Kraemer et al. 1999; Häkkinen et al. 2001),suggesting age-dependent differences in the IGF-Iaxis response to strength training.

The training-induced increase in muscle mass,despite the circulating decrease in IGF-I level, sug-gest that the local tissue effect of exercise on growthfactors differ from systemic effects. Very few studieshave examined the effect of brief exercise or trainingon skeletal muscle IGF-I levels. Yan et al. (1993)demonstrated that an acute bout of eccentric exer-cise led to an increase in IGF-I immunoreactivity inrat type II muscle 4 days post-exercise. Consistentwith Yan’s observation, we found that 5 days oftreadmill training in young female rats resulted in a significant increase in muscle size and muscle IGF-I protein without changes in IGF-I mRNA and

IL-1

*

*

*

IL-6 TNF

ControlTrained

80

60

40

20

0

–20

–40

Ch

ang

e (%

)

Fig. 13.4 The effect of 5-week aerobic-type exerciseintervention on proinflammatory cytokines (interleukin-1β, interleukin-6 and tumor necrosis factor-α). The stressof training was associated with a significant increase of theproinflammatory cytokines. IL, interleukin; TNF, tumornecrosis factor.

Page 190: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

174 chapter 13

circulating IGF-I (Eliakim et al. 1997). These results suggest that the early mechanisms of the trainingadaptation involve translational or post-translationalincreases in muscle tissue IGF-I, and that local muscle IGF-I regulation may be dissociated fromcentral control mechanisms. Along these lines,Phillip et al. (1994) suggested that increases in IGF-Iin the kidney was not due to an increase in local synthesis but rather to an increase in IGF-I uptakefrom the circulation by a non-membrane associatedIGFBP-1. This observation emphasizes the possiblerole of local IGFBPs (which are strongly attached tothe cell surface by a membrane integrin-bindingdomain) in the regulation of the early adaptation oflocal IGF-I to exercise training.

Nevertheless, compensatory hypertrophy of theplantaris and soleus muscle after unilateral excisionof the gastrocnemius tendon was accompanied by asignificant increase in IGF-I mRNA after only 2 days(levels of IGF-I protein were not measured in thesestudies) (De Vol et al. 1990). This suggests that dif-ferent types and amount of muscular work couldlead to a different time course and pattern of localIGF-I increase.

Longer periods of exercise training do, however,lead to stimulation of IGF-I gene expression.Zanconato et al. (1994) found an increase in hepaticIGF-I gene expression following 4 weeks of endur-ance training in young rats. In addition, the authorsshowed that the 4 weeks of endurance type trainingled to increases in the exercising muscle IGF-I geneexpression and protein. Interestingly, inhibition ofGH (by hypophysectomy [De Vol et al. 1990] or byGH releasing hormone antibody [Zanconato et al.1994]) actually enhanced the local IGF-I response toincreased muscular effort. It is clear from theseobservations that inhibition of GH alone cannotblock the autocrine and paracrine effects of IGF-I,emphasizing the GH-independence of the ‘local’IGF-I anabolic adaptations to physical activity.

Singh et al. (1999) studied the effect of progressiveresistance training in frail elderly humans. Theyfound that training was associated with increasedmuscle strength and with muscle fiber remodeling,and that higher baseline muscle IGF-I predicted theincrease in muscle strength.

Recently, it was suggested that there are two

isoforms of IGF-I in the muscle (McKoy et al. 1999;Goldspink & Yang 2001). One of the isoforms isdetectable only after mechanical stimulation andtherefore was named mechano growth factor (MGF).Its response to resistance training is higher in youngadults, indicating age-related desensitivity to mech-anical loading (Hameed et al. 2003). MGF is smallerand unglycosylated, and has a shorter half-life timeand a different receptor binding affinity than thesystemic liver IGF-I. The muscle has another iso-form of IGF-I. This isoform is similar to the liver-type IGF-I, and is also up-regulated by exercise.Induction of MGF expression occurs mainly afterstretch and electrical stimulation. MGF has animportant role in local protein synthesis and in theprevention of apoptosis and, therefore, plays a pivotal role in local tissue repair and remodeling.This emphasizes again the significant importance ofthe local IGF-I response to exercise training.

What is the advantage to the organism of simultan-eous central catabolism and local anabolism early inthe adaptation to increased physical activity? We speculate that this adaptive mechanism mightreduce global anabolic function, thereby conservingenergy sources, but still allows for local tissuegrowth in response to environmental stresses likeexercise training.

Consistent with this speculation is the phe-nomenon of attenuated somatic growth and re-duced circulating IGF-I despite muscle adaptationto intense exercise training in nutritionally self-deprived young elite athletes (e.g. female gymnasts[Theintz et al. 1993]). The dissociation between target tissue and central neuroendocrine GH–IGF-Iresponses was also demonstrated in other environ-mental conditions (Moromisato et al. 1996). Ratsexposed to hypoxia had a reduced growth rate and circulating IGF-I, but the relative size of theirheart and lung was increased along with local IGF-Igene expression. This indicates that a local anabolicadjustment to reduced oxygen carrying capacityhad occurred but that the central response wascatabolic and overall growth was reduced.

In summary, there are differences between thelocal and systemic GH–IGF-I response to exercisetraining (Fig. 13.5). The local and more importantskeletal muscle response is anabolic from very early

Page 191: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 175

stages, and result mainly from GH-independentautocrine and paracrine IGF-I release. The systemicGH–IGF-I response to a training program has atleast two phases: the first is an acute catabolic-typeresponse with a decrease in circulating IGF-I. Atsome later point, depending probably also on thenutritional and energy balance of the individual, achronic anabolic adjustment of the GH–IGF-I axisoccurs. Whether the initial catabolic phase is neces-sary for the later anabolic adaptation, and whetherthis response is affected by the fitness level of theindividual is still unknown.

Possible practical applications for the coach and athlete

The effectiveness of physical training dependsessentially on the intensity, volume, duration andfrequency of training, and on the individual abilityto tolerate training. An imbalance between the train-ing load and the individual’s tolerance leads tounder or overtraining. Therefore, many efforts havebeen made to find objective parameters to quantifythe balance between training load and the athlete’stolerance, with limited success. The endocrine system, by modulation of anabolic and catabolicprocesses, plays a major role in the physiolo-gical adaptation to exercise training (Urhausen &

Kindermann 2000). For example, the change in the testosterone/cortisol ratio, as an indicator of the anabolic–catabolic balance, has been used withlimited success to determine the physiologicalstrain of training (Kuoppasalmi & Adlercreutz 1984;Hoffman et al. 1997). The data presented here sug-gests that changes in circulating IGF-I may alsoserve as a possible marker of the training load, andthat sustained reduced circulating IGF-I levels mayindicate overtraining. This is particularly importantin young competitive athletes, and particularly insports that typically combine intensive trainingwith caloric restriction (aesthetic sports [e.g. gym-nastics] or weight-categories sports [e.g. wrestling]).Reduced circulating IGF-I level in these circum-stances indicates negative energy balance, whichmay lead to attenuated somatic growth (Jahreis et al.1991; Theintz et al. 1993; Roemmich & Sinning 1997).Therefore circulating IGF-I levels can serve as a veryimportant alerting sign, and the athletes should beaware that even when they reduce the trainingintensity, they would not be always able to com-pensate for the growth loss during long periods ofnegative energy balance.

Measurements of IGF-I levels can also help theathlete and coach in the preparation for competi-tion. We recently determined the effect of 4 weeks of training on fitness, self-assessment physical con-ditioning scores and circulating IGF-I in elite profes-sional handball players during their preparation forthe junior world championships (Eliakim et al.2002). Training consisted of 2 weeks of intense train-ing followed by 2 weeks of relative tapering. Bothcirculating IGF-I and physical conditioning scoresdecreased initially, and returned to baseline levelsat the end of training. We found a significant posit-ive correlation between the changes in circulatingIGF-I and the physical conditioning scores.

Tapering down the training intensity prior to thecompetition is a well-known training methodologyto help the athlete to achieve his best performance.The results of this study demonstrated that thisstrategy is indeed associated with parallel changesin both IGF-I (an objective measure) and in indi-vidual conditioning self-assessment (a subjectivemeasure). Therefore, both measures may assistcoaches and athletes in their training preparations.

Systemic response

Local response

Baseline levels

Training duration

IGF-

I

Fig. 13.5 Differences between local and systemicadaptations of insulin-like growth factor I (IGF-I) toexercise training. While muscle IGF-I increases from veryearly stages of training, there are at least two phases in the systemic IGF-I response. The first phase is an acutecatabolic-type response characterized by decreases incirculating IGF-I, but at some point anabolic reboundoccurs and IGF-I increases.

Page 192: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

176 chapter 13

However, as noted earlier (e.g. Eliakim et al. 1996,2001), despite the decrease in circulating IGF-I,fitness improved. These results suggest that whilechanges in circulating IGF-I are good markers of thegeneral condition and energy balance of the athlete,they are not necessarily good predictors of the athlete’s performance.

Recently, we measured IGF-I level throughoutthe training season in elite judokas. IGF-I decreasedsignificantly during periods of heavy training, butreturned to baseline levels during tapering down,and reached a peak above baseline levels during thecompetition period (Fig. 13.6) (unpublished data).Nemet et al. (2004) showed similar patterns for free IGF-I during the training season in competitivewrestlers. It is still unknown what the permitteddecrease of IGF-I during periods of heavy trainingis, or the optimal increase during periods of taper-ing down and reduced training intensity. However,we believe that the inability to increase circulatingIGF-I levels before the target competition should be an alarming sign for both the athlete and his/hercoach that the athlete’s general condition is not optimal.

Finally, we demonstrated that local IGF-I changesare more important than systemic changes in themuscle adaptation to exercise training. However, if indeed GH changes following exercise play a keyrole in the anabolic effects of exercise, attention

should be paid by athletes and trainers to factorsthat may inhibit the GH response to exercise andattenuate the training effect (e.g. low exercise intens-ity; short intervals between multiple daily exercisesessions; exercise following high fat meal; continu-ous training despite menstrual irregularities, etc.).

Pres

easo

n0

Light–

moder

ate

train

ing

Heavy

train

ing

Tapar

ing

down

Compet

ition

420 400 380 360 340 320 300 280 260 240 220 200

IGF-

I (n

g·m

L–1)

Fig. 13.6 A typical example of insulin-like growth factor I(IGF-I) levels in elite judoka during the training season.IGF-I level decreased significantly during heavy training,and increased above baseline levels before the targetcompetition.

ReferencesBamman, M.M., Shipp, J.R., Jiang, J. et al.

(2001) Mechanical load increases muscleIGF-I and androgen receptor mRNAconcentrations in humans. AmericanJournal of Physiology 280, E383–E390.

Bang, P., Brandt, J., Degerblad, M. et al.(1990) Exercise-induced changes ininsulin-like growth factors and their lowmolecular weight binding proteins inhealthy subjects and patients withgrowth hormone deficiency. EuropeanJournal of Clinical Investigation 20,285–292.

Bar-Or, O. (1983) Growth hormonedeficiencyausing exercise in thediagnosis. In: Pediatric Sports Medicine forthe Practitioner (Bar-Or, O., ed.).Springer-Verlag, New York: 182–191.

Baumann, G., Shaw, M.A. & Amburn, K.(1989) Regulation of plasma growth

hormone binding proteins in health anddisease. Metabolism 38, 683–689.

Berelowitz, M., Szabo, M., Frohman, L.A.et al. (1981) Somatomedin-C mediatesgrowth hormone negative feedback byeffects on both the hypothalamus andthe pituitary. Science 212, 1281–1297.

Bermon, S., Ferrari, P., Bernard, P., Altare,S. & Dolisi, C. (1999) Responses of totaland free insulin-like growth factor-I andinsulin-like growth factor-I bindingprotein-3 after resistance exercise andtraining in elderly subjects. ActaPhysiologica Scandinavica 165, 51–56.

Borer, K.T., Nicolski, D.R. & Owens, V.(1986) Alteration of pulsatile growthhormone secretion by growth inducingexercise: involvement of endogenousopiates and somatostatin. Endocrinology118, 844–850.

Borst, S.E., De Hoyos, D.V., Garzarella, L.et al. (2001) Effects of resistance trainingon insulin-like growth factor and IGFbinding proteins. Medicine and Science inSports and Exercise 33, 648–653.

Bouchard, C. & Despres, J.P. (1989)Variation in fat distribution with ageand health implications. In: PhysicalActivity and Aging (Spirduso, W.W. &Eckert, H.M., eds.). Human Kinetics,Champaign, IL: 78–106.

Buckler, J.M.H. (1972) Exercise as ascreening test for growth hormonerelease. Acta Endocrinologica 69, 219–229.

Buckler, J.M.H. (1973) The relationshipbetween changes in plasma growthhormone levels and body temperatureoccurring with exercise in man.Biomedicine 19, 193–197.

Cappon, J.P., Ipp, E., Brasel, J.A. & Cooper,

Page 193: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 177

D.M. (1993) Acute effects of high-fat andhigh-glucose meals on the growthhormone response to exercise. Journal ofClinical Endocrinology and Metabolism 76,1418–1422.

Cappon, J.P., Brasel, J.A., Mohan, S. &Cooper, D.M. (1994) Effect of briefexercise on circulating insulin-likegrowth factor-I. Journal of AppliedPhysiology 76, 2490–2496.

Casanueva, F.F., Villanueva, L., Cabezas, J.& Fernandez-Cruz, A. (1984)Cholinergic mediation of growthhormone secretion elicited by arginineclonidine and physical exercise in man.Journal of Clinical Endocrinology andMetabolism 59, 526–532.

Casanueva, F.F., Villanueva, L., Dieguez,C. et al. (1987) Free fatty acids blockgrowth hormone (GH) releasinghormone stimulated GH secretion inman directly at the pituitary. Journal ofClinical Endocrinology and Metabolism 65,634–642.

Chadan, S.G., Dill, R.P., Vanderhoek, K. & Parkhouse, W.S. (1999) Influence ofphysical activity on plasma insulin-likegrowth factor-I and insulin-like growthfactor-I binding proteins in healthy olderwomen. Mechanisms of Ageing andDevelopment 109, 21–34.

Clark, R.G., Jansson, J.O., Isaksson, O. & Robinson, I.C. (1985) Intravenousgrowth hormone: growth responses to patterned infusions inhypophysectomized rats. Journal ofEndocrinology 104, 53–61.

Convertino, V.A., Keil, L.C., Bernauer,E.M. & Greenleaf, J.E. (1981) Plasmavolume, osmolality, vasopressin andrenin activity durin graded exercise inman. Journal of Applied Physiology 50,123–128.

Corpas, E., Harman, S.M. & Blackman,M.R. (1993) Human growth hormoneand human aging. Endocrine Reviews 14,20–39.

Cowell, C.T. (1995) Short stature. In:Clinical Paediatric Endocrinology, 3rd edn.(Brook C.G.D., ed.). Blackwell Science,Oxford, UK: 136–172.

Dall, R., Lange, K.H., Kjaer, M. et al. (2001)No evidence of insulin-like growthfactor binding protein-3 proteolysisduring a maximal exercise test inathletes. Journal of Clinical Endocrinologyand Metabolism 86, 669–674.

De Palo, E.F., Gatti, R., Lancerin, F. et al.(2002) Urinary insulin-like growthfactor-I in athletes before and afterphysical exercise, and in sedentarysubjects. Clinica Chimica Acta 322, 51–57.

Felsing, N.E., Brasel, J.A. & Cooper, D.M.(1992) Effect of low and high intensityexercise on circulating growth hormonein men. Journal of Clinical Endocrinologyand Metabolism 75, 157–162.

Flamm, S.D., Taki, J., Moore, R. et al. (1990)Redistribution of regional and organblood volume and effect of cardiacfunction in relation to upright exerciseintensity in healthy human subjects.Circulation 81, 1550–1559.

Fraiser, S.D. (1974) A review of growthhormone stimulation tests in children.Pediatrics 53, 929–937.

Goldspink, G. & Yang, S.Y. (2001) Effects of activity on growth factor expression.International Journal of Sport and ExerciseMetabolism 11 (suppl.), 21–27.

Häkkinen, K., Pakarinen, A., Kraemer, W.J. et al. (2001) Selective musclehypertrophy, changes in EMG and force,and serum hormones during strengthtraining in older women. Journal ofApplied Physiology 91, 569–580.

Hameed, M., Orrell, R.W., Cobbold, M.,Goldspink, G. & Harridge, S.D. (2003)Expression of IGF-I splice variants inyoung and old human skeletal muscleafter high resistance exercise. Journal ofPhysiology 547, 247–254.

Hartley, L.H., Mason, J.W., Hogan, R.P. et al. (1972) Multiple hormonalresponses to graded exercise in relationto physical training. Journal of AppliedPhysiology 33, 602–606.

Ho, K.Y., Evans, W.S. & Blizzard, R.M.(1987) Effects of sex and age on 24 hprofile of growth hormone secretion in man: importance of endogenousedtradiol concentration. Journal ofClinical Endocrinology and Metabolism 64,51–58.

Hoffman, J.R., Falk, B., Radom Isaac, S. et al. (1997) The effect of environmentaltemperature on testosterone and cortisolresponses to high intensity, intermittentexercise in humans. European Journal ofApplied Physiology 75, 83–87.

Hopkins, N.J., Jakeman, P.M., CwyfanHughes, S.C. & Holly, J.M.P. (1994)Changes in circulating insulin-likegrowth factor binding protein-1 during prolonged exercise: effect ofcarbohydrate feeding. Journal of ClinicalEndocrinology and Metabolism 79,1887–1890.

Jahreis, G., Kauf, E., Frohner, G. &Schmidt, H.E. (1991) Influence ofintensive exercise on insulin-like growthfactor-I, thyroid and steroid hormones infemale gymnasts. Growth Regulation 1,95–99.

DeVol, D.L., Rotwein, P., Sadow, J.L.,Novakofski, J. & Bechtel, P.J. (1990)Activation of insulin-like growth factorgene expression during work-inducedskeletal muscle growth. American Journalof Physiology 259, E89–E95.

Eliakim, A., Brasel, J.A., Mohan, S. et al.(1996) Physical fitness, endurancetraining, and the GH–IGF-I system inadolescent females. Journal of ClinicalEndocrinology and Metabolism 81,3986–3992.

Eliakim, A., Moromisato, M.Y.,Moromisato, D.Y. et al. (1997) Increase in muscle IGF-I protein but not IGF-ImRNA after 5 days of endurancetraining in young rats. American Journalof Physiology 273, R1557–R1561.

Eliakim, A., Brasel, J.A., Barstow, T.J.,Mohan, S. & Cooper, D.M. (1998a)Fitness and the growth hormone–insulin-like growth factor-I axis inadolescent males. Medicine and Science in Sports and Exercise 30, 512–517.

Eliakim, A., Brasel, J.A., Mohan, S., Wong,W.L.T. & Cooper, D.M. (1998b) Inceasedphysical activity and the growthhormone–IGF-I axis in adolescent males.American Journal of Physiology 275,R308–R314.

Eliakim, A., Brasel, J.A. & Cooper, D.M.(1999) GH response to exercise:assessment of the pituitary refractoryperiod, and relationship with circulating components of the GH–IGF-Iaxis in adolescent females. Journal ofPediatric Endocrinology and Metabolism 12,47–55.

Eliakim, A., Oh, Y. & Cooper, D.M. (2000)Effect of unilateral wrist flexion exerciseon circulating growth hormone, insulin-like growth factor-I, and fibroblastgrowth factor-2. American Journal ofPhysiology 279, R548–R553.

Eliakim, A., Scheet, T.P., Newcomb, R.,Mohan, S. & Cooper, D.M. (2001)Fitness, training and the growthhormone–insulin-like growth factor-Iaxis in prepubertal girls. Journal ofClinical Endocrinology and Metabolism 86,2797–2802.

Eliakim, A., Nemet, D., Bar-Sela, S., Higer, Y. & Falk, B. (2002) Changes incirculating IGF-I and their correlationwith self-assessment and fitness amongelite athletes. International Journal ofSports Medicine 23, 600–603.

Elias, A.N., Pandian, M.R., Wang, L. et al. (2000) Leptin and IGF-I levels inunconditioned male volunteers aftershort term exercise.Psychoneuroendocrinology 25, 453–461.

Page 194: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

178 chapter 13

Jorgensen, J.O., Pedersen, S.B., Borglum, J.et al. (1995) Serum concentration ofinsulin-like growth factors, IGF bindingproteins 1 and 3, and growth hormonebinding proteins in obese women and the effect of growth hormoneadministration: a double blind, placebo-controlled study. European Journal ofEndocrinology 133, 65–70.

Kanaley, J.A., Weltman, J.Y., Veldhuis, J.D.et al. (1997) Human growth hormoneresponse to repeated bouts of aerobicexercise. Journal of Applied Physiology 83,1756–1761.

Kelijman, M. & Frohman, L.A. (1991) Therole of the cholinergic pathway ingrowth hormone feedback. Journal ofClinical Endocrinology and Metabolism 72,1081–1087.

Kelley, P.J., Eisman, J.A., Stuart, M.C. et al.(1990) Somatomedin-C, physical fitness,and bone density. Journal of ClinicalEndocrinology and Metabolism 70,718–723.

Koziris, L.P., Hickson, R.C., Chatterton,R.T. et al. (1999) Serum levels of total andfree IGF-I and IGFBP-3 are increasedand maintained in long-term training.Journal of Applied Physiology 86,1436–1442.

Kraemer, W.J., Häkkinen, K., Newton, R.U.et al. (1999) Effects of heavy-resistancetraining on hormonal response patternsin younger versus older men. Journal ofApplied Physiology 87, 982–992.

Kraemer, W.J., Harman, F.S., Vos, N.H. etal. (2000) Effects of exercise and alkalosison serum insulin-like growth factor-Iand IGF binding protein-3. CanadianJournal of Applied Physiology 25, 127–138.

Kratzsch, J., Blum, W.F., Ventz, M. et al.(1995) Growth hormone binding proteinrelated immunoreactivity in the serumof patients with acromegaly is regulatedinversely by growth hormoneconcentration. European Journal ofEndocrinology 132, 306–312.

Kuoppasalmi, K. & Adlercreutz, H. (1984)Interaction between catabolic andanabolic steroid hormones in muscularexercise. In: Exercise Endocrinology(Fotherby, K. & Pal, S.B., eds.). DeGruyter, Berlin: 65–98.

Lamson, G., Giudice, L.C., Cohen, P. et al.(1993) Proteolysis of IGFBP-3 may be acommon regulatory mechanism of IGFaction in vivo. Growth Regulation 3, 91–95.

Leblanc, A.D., Schneider, V.S., Evans, H.J.,Engelbretson, D.A. & Krebs, J.M. (1990)Bone mineral loss and recovery after 17weeks of bed rest. Journal of Bone MineralResearch 5, 843–850.

Mohan, S., Nakao, Y., Honda, Y. et al.(1995) Studies on the mechanisms bywhich insulin-like growth factor (IGF)binding protein-4 (IGFBP-4) and IGFBP-5 modulate IGF actions in bone cells.Journal of Biological Chemistry 270, 20 424–20 431.

Moromisato, D.Y., Moromisato, M.Y.,Zanconato, S. et al. (1996) Effect ofhypoxia on lung, heart, and liver insulin-like growth factor-I gene and receptorexpression in the newborn rat. CriticalCare Medicine 24, 919–924.

Nemet, D., Oh, Y., Kim, H.S., Hill, M. &Cooper, D.M. (2002) Effect of intenseexercise on inflammatory cytokines andgrowth mediators in adolescent boys.Pediatrics 110, 681–689.

Nemet, D., Connolly, P.H., Pontello-Pescatello, A.M. et al. (2004) Negativeenergy balance plays a major role in the IGF-I response to exercise training.Journal of Applied Physiology 96, 276–282.

Nemet, D., Pontello, A.M., Rose-Gottron,C. & Cooper, D.M. (2004) Cytokines and growth factors during and after awrestling season in adolescent boys.Medicine and Science in Sports and Exercise36, 794–800.

Nindl, B.C., Kraemer, W.J., Marx, J.O. et al. (2001) Overnight responses of thecirculating IGF-I system after acuteheavy resistance exercise. Journal ofApplied Physiology 90, 1319–1326.

Phillip, M., Werner, H., Palese, T. et al.(1994) Differential accumulation ofinsulin-like growth factor-I in kidneys of pre- and postpubertal streptozotocin-diabetic rats. Journal of MolecularEndocrinology 12, 215–224.

Poehlman, E.T. & Copeland, K.C. (1990)Influence of physical activity on insulin-like growth factor-I in healthy youngerand older men. Journal of ClinicalEndocrinology and Metabolism 70,1468–1473.

Pontiroli, A.E., Lanzi, R., Monti, L.D.,Sandoli, E. & Pozza, G. (1991) Growthhormone (GH) autofeedback on GHresponse to GH-releasing hormone. Role of free fatty acids and somatostatin.Journal of Clinical Endocrinology andMetabolism 72, 492–495.

Postel-Vinay, M.C., Saab, C. & Gourmelen,M. (1995) Nutritional status and growthhormone binding protein. HormoneResearch 44, 177–181.

Raastad, T., Bjoro, T. & Hallen, J. (2000)Hormonal responses to high andmoderate intensity strength exercise.European Journal of Applied Physiology 82,121–128.

Lee, C., Eliakim, A., Brasel, J.A. & Cooper,D.M. (2000) Effects of exercise trainingon red blood cell insulin-like growthfactor-I receptor binding in adolescentmales. Journal of Pediatric Endocrinologyand Metabolism 13, 621–627.

Leung, D.W., Spencer, S.A., Cachaines, G.et al. (1987) Growth hormone receptorand serum binding proteins:purification, cloning, and expression.Nature 330, 537–540.

Lowe, W.L., Jr. (1991) Biological actions of the insulin-like growth factors. In:Insulin-like Growth Factors: Molecular andCellular Aspects (LeRoith, D., ed.). CRC,Boca Raton, FL: 49–86.

Manetta, J., Brun, J.F., Maimoun, L. et al.(2002) Effect of training on the GH/IGF-Iaxis during exercise in middle-agedmen: relationship to glucosehomeostasis. American Journal ofPhysiology 283, E929–E936.

Marcus, R.G., Buttterfield, G., Holloway, L. et al. (1990) Effects of short-termadministration of recombinant humangrowth hormone to elderly people.Journal of Clinical Endocrinology andMetabolism 70, 519–527.

Marimme, T.J., Zapf, J. & Froesch, E.R.(1982) Insulin-like growth factors in thefed and fasted states. Journal of ClinicalEndocrinology and Metabolism 55,999–1002.

Martin, J.L. & Baxter, R.C. (1986) Insulin-like growth factor binding protein fromhuman plasma. Journal of BiologicalChemistry 261, 8754–8760.

Mauras, N., Blizzard, R.M., Link, K. et al.(1987) Augmentation of growthhormone secretion during puberty:evidence for a pulse amplitude-modulated phenomenon. Journal ofClinical Endocrinology and Metabolism 64,596–601.

McCall, G.E., Byrnes, W.C., Fleck, S.J.,Dickinson, A. & Kraemer, W.J. (1999)Acute and chronic hormonal responsesto resistance training designed topromote muscle hypertrophy. CanadianJournal of Applied Physiology 24, 96–107.

McKoy, G., Ashley, W., Mander, J. et al.(1999) Expression of insulin-like growthfactor-I splice variants and structuralgenes in rabbit skeletal muscle inducedby stretch and stimulation. Journal ofPhysiology 516, 583–592.

Mohan, S. & Baylink, D.J. (1991) The role of insulin-like growth factor-II in the coupling of bone formation toresorption. In: Modern Concepts ofInsulin-like Growth Factors (Spencer,E.M., ed.). Elsevier, New York: 169–184.

Page 195: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise, training and the gh– igf-i axis 179

Raastad, T., Glomsheller, T., Bjoro, T. &Hallen, J. (2001) Changes in humanskeletal muscle contractility andhormonal status during 2 weeks ofheavy strength training. European Journalof Applied Physiology 84, 54–63.

Rajaram, S., Baylink, D.J. & Mohan, S.(1997) Insulin-like growth factor bindingproteins in serum and other biologicalfluids: regulation and function.Endocrine Reviews 18, 801–831.

Roemmich, J.N. & Sinning, W.E. (1997)Weight loss and wrestling training:effects on growth-related hormones.Journal of Applied Physiology 82,1760–1764.

Ronsen, O., Haug, E., Pedersen, B.K. & Bahr, R. (2001) Increasedneuroendocrine response to a repeatedbout of endurance exercise. Medicine and Science in Sports and Exercise 33,568–575.

Rosenfeld, R.G. (1994) Circulating growthhormone binding proteins. HormoneResearch 42, 129–132.

Rosenfeld, R.G., Albertsson-Wikland, K.,Cassorla, F. et al. (1995) Diagnosticcontroversy: the diagnosis of childhoodgrowth hormone deficiency revisited.Journal of Clinical Endocrinology andMetabolism 80, 1532–1540.

Scheet, T.P., Mills, P.J., Ziegler, M.G.,Stoppani, J. & Cooper, D.M. (1999) Effect of exercise on cytokines andgrowth mediators in prepubertalchildren. Pediatric Research 46, 429–434.

Scheet, T.P., Nemet, D., Stoppani, J. et al.(2002) The effect of endurance-typetraining on growth mediators andinflammatory cytokines in prepubertaland early pubertal males. PediatricResearch 52, 491–497.

Schwarz, A.J., Brasel, J.A., Hintz, R.L.,Mohan, S. & Cooper, D.M. (1996) Acuteeffect of brief low and high intensityexercise on circulating IGF-I, II, and IGF-I binding protein-3 and its proteolysis inyoung healthy men. Journal of ClinicalEndocrinology and Metabolism 81,3492–3497.

Waters, D.L., Qualls, C.R., Dorin, R.,Veldhuis, J.D. & Baumgartner, R.N.(2001) Increased pulsatility, processirregularity, and nocturnal troughconcentrations of growth hormone inamenorrheic compared to eumenorrheicathletes. Journal of Clinical Endocrinologyand Metabolism 86, 1013–1019.

Weltman, A., Weltman, J.Y., Schurrer, R. et al. (1992) Endurance training amplifiesthe pulsatile release of growth hormone:effects of training intensity. Journal ofApplied Physiology 72, 2188–2196.

Weltman, A., Weltman, J.Y., Hartman,M.L. et al. (1994) Relationship betweenage, percentage body fat, fitness, and 24-hour growth hormone release in healthyyoung adults: effects of gender. Journal ofClinical Endocrinology and Metabolism 78,543–548.

Wideman, L., Weltman, J.Y., Shah, N. et al.(1999) Effects of gender on exercise-induced growth hormone release. Journalof Applied Physiology 87, 1154–1162.

Wilkinson, P.W. & Parkin, J.M. (1974)Growth hormone response to exercise in obese children. Lancet 2, 55.

Yamashita, S. & Melmed, S. (1986) Effectsof insulin on rat anterior pituitary cells:inhibition of growth hormone secretionand mRNA levels. Diabetes 35, 440–447.

Yan, Z., Biggs, R.B. & Booth, F.W. (1993) Insulin-like growth factorimmunoreactivity increases in muscleafter acute eccentric contractions. Journalof Applied Physiology 74, 410–414.

Zaccaria, M., Varnier, M., Piazza, P.,Noventa, D. & Ermolao, A. (1999)Blunted growth hormone response to maximal exercise in middle-agedversus young subjects and no effect ofendurance training. Journal of ClinicalEndocrinology and Metabolism 84,2303–2307.

Zanconato, S., Moromisato, D.Y.,Moromisato, M.Y. et al. (1994) Effects of training and growth hormonesuppression on insulin-like growthfactor-I mRNA in young rats. Journal ofApplied Physiology 76, 2204–2209.

Singh, M.A., Ding, W., Manfredi, T.J. et al.(1999) Insulin-like growth factor-I inskeletal muscle after weight-liftingexercise in frail elders. American Journalof Physiology 277, E135–E143.

Smith, A.T., Clemmons, D.R., Underwood,L.E., Ben Ezra, V. & McMurray, R. (1987) The effect of exercise on plasmasomatomedin-C/insulin-like growthfactor I concentrations. Metabolism 36,533–537.

Smith, W.J., Underwood, L.E. &Clemmons, D.R. (1995) Effects of caloricand protein restriction on insulin-likegrowth factor-I (IGF-I) and IGF-bindingproteins in children and adults. Journal ofClinical Endocrinology and Metabolism 80,443–449.

Suikkary, A.M., Sane, T., Seppala, M. et al.(1989) Prolonged exercise increasesserum insulin-like growth factor bindingprotein concentrations. Journal of ClinicalEndocrinology and Metabolism 68,141–144.

Sutton, J.R. & Lazarus, L. (1976) Growthhormone and exercise comparison ofphysiological and pharmacologicalstimuli. Journal of Applied Physiology 41,523–527.

Theintz, G.E., Howald, H., Weiss, U. &Sizonenko, P.C. (1993) Evidence for areduction of growth potential inadolescent female gymnasts. Journal ofPediatrics 122, 306–313.

Tonshoff, B., Blum, W.F., Wingen, A.M. & Mehls, O. (1995) Serum insulin-likegrowth factors (IGFs) and IGF bindingproteins 1, 2 and 3 in children withchronic renal failure: relationship toheight and glomerular filtration rate.The European Study Group forNutritional Treatment of Chronic RenalFailure in Childhood. Journal of ClinicalEndocrinology and Metabolism 80,2684–2691.

Urhausen, A. & Kindermann, W. (2000)The endocrine system in overtraining.In: Sports Endocrinology (Warren, M.P. &Constantini, N.W., eds.). Humana Press,Totowa, NJ: 347–370.

Page 196: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

180

Introduction

Molecular biology methods have drastically changedthe way we think about hormones. The classicaldefinition of a hormone was ‘a chemical substanceproduced in a specialized gland released into theblood stream and transported to different tissue toelicit a physiological purpose’. We are now awarethat hormones, or factors, are produced by manydifferent types of tissues, and that some have anautocrine or a paracrine effect. This is exemplifiedby the insulin-like growth factor I (IGF-I) system, afamily of different forms of IGF-I, each of which hasa different biological effect. These are derived fromthe splicing of the IGF-I gene in response to differentsignals. The human genome has now been sequencedand comprises about 40 000 different genes. How-ever, we know there are many more different pro-teins, so it is therefore evident that the same genemust be spliced differentially to generate this pheno-typic diversity. The IGF gene appears to have evolvedfrom a single insulin-like gene that is expressed invertebrates. A similar insulin-like gene is present inthe C. elegans, the nematode, and in the chordateamphioxus, closely related to the common ancestorof the vertebrate. During vertebrate evolution how-ever, the gene has been duplicated to give rise toinsulin, IGF-I and IGF-II genes. Experiments on C.elegans have shown that the ancestral insulin-likegene prevents cell death and considerably extendsthe life of the worm. The system is even more versatilebecause the insulin-like growth genes can be splicedto produce different RNA transcripts and differentpolypeptides with different biological activity.

IGF-I, originally called ‘somatomedin’, was re-garded as a general growth factor produced by theliver under the influence of growth hormone (GH).Later, it became apparent that it is expressed bymost tissues and exists as different splice variants,each of which has a somewhat different action.

Growth hormone–IGF-I axis

The original somatomedin hypothesis originated inthe 1950s following early efforts to understand theregulation of somatic growth by pituitary-derivedGH. It was suggested that this did not act directly on its target tissues to promote growth, but therewere intermediary substances involved (Daughaday& Reeder 1966). The term ‘somatomedin’ was lateradopted (Daughaday et al. 1972) to reflect the growthpromoting actions of these substances, which weresubsequently characterized and later called insulin-like growth factors (Rinderknecht & Humbel 1978;Klapper et al. 1983). However, in 1985, Green et al.proposed the ‘dual effector hypothesis’ (Green et al.1985), which suggested that GH had direct effectson peripheral tissues that were not mediated byIGF-I and that GH stimulated local IGF-I produc-tion. It is now clear that one of its main roles is stimu-lating the release of IGF-I from the liver and that, inaddition, GH stimulates the formation of a ternaryIGF binding complex, including insulin-like growthfactor binding protein 3 (IGFBP-3) and the acid-labile subunit (ALS), which stabilizes IGF-I in theserum. GH is secreted from somatotroph cells locatedwithin the anterior pituitary gland in a pulsatilemanner, and also has itself other specific functional

Chapter 14

The Role of MGF and Other IGF-I Splice Variants inMuscle Maintenance and Hypertrophy

GEOFFREY GOLDSPINK, SHI YU YANG, MAHJABEEN HAMEED,

Page 197: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 181

effects on local tissues. These include a proteinanabolic effect (increased DNA, RNA and proteinsynthesis), stimulation of growth and calcificationof cartilage, an increased mobilization of fats, anduse of fats as an energy source. The exercise-inducedincrease in GH is thought to mediate, either directlyor indirectly, many of the somatotrophic events sur-rounding tissue remodeling (reviewed by Nindl et al. 2003).

Unquestionably, the GH–IGF-I axis plays a role in postnatal growth and development, and thesehormones reach their peak during adolescence.However, with increasing age, a further decline inthe circulating levels of GH and IGF-I occurs, suchthat older people can be regarded as partially GHdeficient (Rudman et al. 1981). The relationshipbetween the age-related decline in the GH–IGF-Iaxis and loss of muscle mass and strength has beenextensively studied. In young GH-deficient adults,administration of recombinant human growth hor-mone (rhGH) was shown to have positive effects onmuscle mass and function (Cuneo et al. 1991).Another study where GH-deficient adults weretreated with GH for an extended period of time con-cluded that there was not only increased musclestrength but also decreased body fat (Beshyah et al.1995). This led to the belief that older individualswith decreased levels of circulating GH and IGF-Iwould also benefit from rhGH therapy. However,studies which have combined GH administrationand resistance training in both young (Yarasheski et al. 1992) and older men (Yarasheski et al. 1995)have shown that the rates of protein synthesis are no greater when resistance training is combinedwith GH than when resistance training is performedalone. Furthermore, in older people the changes inmuscle mass and function have also been reportedto be similar between both groups (Lange et al.2002). It should be noted that studies involving theuse of rhGH, such as those listed above, are likely tohave used the 22 kDa molecular isoform of GH. Thisis the predominant form of GH found in the plasmaGH. The effects of the other molecular isoforms ofGH, of which there are thought to be more than ahundred (Baumann, 1991), are yet to be determined.The roles of circulating GH and IGF-I, particularlywith regard to muscle adaptation in later life, are

still unclear. Systemic growth factors may be of rel-atively minor importance in muscle hypertrophy.For example, in one study the overloaded muscles of hypophysectomized rats were still able to hyper-trophy despite significantly reduced systemic IGF-Ilevels (Adams & Haddad 1996). These findings,coupled with the simple observation that it is onlychallenged muscles that hypertrophy and not all themuscles of the body, highlights the importance of a‘local’ system of muscle adaptation.

Expression of IGF-I splice variants in muscle

Muscles can be stimulated to grow rapidly ifmechanically challenged; electrical stimulation of amuscle held in a stretched position also promotesboth muscle lengthening (by the addition of newsarcomeres in series) and increases cross-sectionalarea, by adding sarcomeres in parallel (Goldspink et al. 1992). Using this approach in combination withspecific primers and RT-PCR (reverse transcription-polymerase chain reaction) it was possible to detecttwo different RNA transcripts (Yang et al. 1996),which subsequent cloning and sequencing identi-fied as being derived from the IGF-I gene by altern-ative splicing (Yang et al. 1996). The first, IGF-IEawas also detected in the resting muscle, and corres-ponded to the transcript commonly expressed in the liver. The second, not detected in resting muscle,corresponded to IGF-IEb.

The terminology of the IGF-I is a problem whenattempting to apply it to non-hepatic tissues andtherefore this splice variant in muscle was namedmechano growth factor (MGF), due to its apparentregulation by mechanical signals (Yang et al. 1996);it has a different carboxy-peptide sequence to theliver type of IGF-I. An additional problem is thatMGF would be classified as IGF-IEb in rat but IGF-IEc (as identified in hepatoma cells by Chew et al.1995) in humans (Table 14.1). Also, in human musclean additional transcript has been detected (Hameedet al. 2004), which confusingly has been termed IGF-IEb but which differs from the rat IGF-IEb. It istherefore apparent that the muscle IGF-I isoformswhile related to the liver isoforms, need to be char-acterized separately (Table 14.1 and Fig. 14.1).

Page 198: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

182 chapter 14

Mechano growth factor

As well as MGF being expressed in response tomechanical activity, it was noted that its E domainhad an insert that changes the open reading frame.Amino acids are encoded by nucleotide base tripletsand any insert that is not a multiple of three there-fore changes the downstream sequence. There is a52 base pair insert in the rat and a 49 base pair insertin humans. This has important functional conse-quences, as the 3’RNA sequence codes for a differ-ent carboxy peptide sequence that is involved in the recognition of the binding proteins. Also, in thecase of MGF, the carboxy peptide (encoded in exons5 and 6) sequence acts as a different growth factor tothe peptide that binds to the IGF-I receptor (encodedin exons 3 and 4). The E-domain peptide alone hasbeen shown to induce division of mononucleatedmyoblasts and thus activate the muscle satellite(stem) cells required for muscle hypertrophy andrepair (Yang & Goldspink 2002). MGF is apparentlynot glycosylated and there is evidence that it has a

short half-life unless bound to the binding proteinwhich may be intracellular. Therefore MGF can beregarded as an autocine/paracrine or local growthfactor produced locally in response to mechanicalstimuli and acts on those muscle fibers that produceit. Hence it is an important signaling molecule in thelocal regulation of muscle growth.

Systemic IGF-I is produced by active muscle

As mentioned above, IGF-IEa is also expressed inskeletal muscle as well as in several other non-hepatic tissues. It has a similar sequence to the mainisoform produced by the liver and is thereforeassumed to have a systemic action. However, muscle expresses at least two of the major bindingproteins of the systemic form of IGF-I and theirexpression tends to be up-regulated as is that of IGF-IEa, by exercise. As the IGF-IEa produced by musclewill bind to these binding proteins in the extracel-lular matrix as well as in the serum, it is expected to have more effect on the muscles that produce itthan on other muscles; its action can be regarded asautocrine and paracrine, as well as endocrine.

As well as having a different carboxy peptidesequence, the expression kinetics are different tothose of MGF. It was shown by Haddad and Adams(2002), that in response to resistance-type exercise in rats, the mRNA of MGF was expressed earlierthan IGF-IEa mRNA. In support of this, Hill andGoldspink (2003) showed that in rats that, followingmuscle damage, MGF is produced as a pulse lastinga few days, whereas the expression of IGF-IEa

Table 14.1 The different names used to describe theinsulin-like growth factor I (IGF-I) isoforms in theliterature.

IGF-I isoform Names used in the literature

IGF-IEa L.IGF-I*, m-IGF-I†

IGF-IEbIGF-IEc IGF-IEb (rat)‡, MGF*

*Yang et al. (1996); †Musaro et al. (2001); ‡Rotwein et al.(1986).MGF, mechano growth factor.

EXONS 1 2 3 4 5 6

Human IGF-I gene

IGF-IEc

IGF-IEb

IGF-IEa

Fig. 14.1 Schematic representation ofthe human IGF-I gene comprising ofsix exons. The three splice variants,IGF-IEa, IGF-IEb and IGF-IEc (alsoknown as mechano growth factor,MGF) expressed in muscle are shownwith the relevant exons. The blackbox in exon 5 represents the first 49base pairs (bp) (52 bp in the rat)which gives rise to the alternativelyspliced, mechano-sensitive MGFisoform.

Page 199: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 183

increases as MGF declines but stays elevated formuch longer.

IGF-I and its splice variants in human muscle

In a study of young military recruits, Hellsten et al.(1996) reported an increase in IGF-I immunoreact-ivity in muscle after 7 days of strenuous exercise(which included terrain marching and warfare exer-cises). More, recently, in a study of elderly people,Singh et al. (1999) reported a 500% increase in IGF-Ilevels in the quadriceps muscles following a 12-week period of resistance training, as determined by immunohistochemistry. Resistance training con-sisted of three sets of eight repetitions at 80% of themost recently determined 1 repetition maximum (1-RM) for the hip and knee extensor muscles, 3 daysper week for 10 weeks. It is clear that this type ofexercise training, where the active muscles mustovercome high loads, is the type of exercise thatresults in muscle hypertrophy. However, thesestudies failed to distinguish between the differentIGF-I splice variants. Recently, the mRNA levels ofMGF and IGF-IEa were measured using real-timequantitative PCR shortly (2.5 h) after a single bout of high intensity knee extensor exercise. Subjects in this study performed 10 sets of six repetitions of the knee extensor muscles at 80% of their 1-RM.(Hameed et al. 2003a). In young subjects it wasobserved that MGF mRNA levels were significantlyincreased as a result of weightlifting exercise, but nosuch change was observed in older subjects. Fur-thermore, at this short time point after exercise IGF-IEa mRNA levels were unchanged in both groups.These observations were interesting in that theywere in general agreement with animal experimentsin which MGF levels were shown to increase beforethose of IGF-IEa, suggesting that the two isoformswere differentially regulated. No relationship wasobserved with muscle myosin heavy chain isoformcomposition, but of note was the observation thatthe subject who showed the most dramatic increasein MGF was the subject whose muscles expressedthe most MHC-IIX. Weightlifting exercise com-prises both concentric and eccentric componentsand in a recent study, Bamman et al. (2001) reporteda 62% increase in IGF-I mRNA concentration in the

muscle, 48 h after an acute bout of eccentric, but notconcentric contractions. Shortly (2.5 h) after a boutof eccentric cycling exercise, we again detected asignificant increase in MGF, but not IGF-IEa. Theeccentric exercise consisted of 60 min of reversepedal cycling divided into six work intervals: 0–6 min at 50%, 6–12 min at 75%, 12–20 min at 100%,20–25 min at 130%, 25–40 min at 100% and 40–60 min at 75% of the load corresponding to the pre-viously determined concentric Vo2max (Hameed et al. 2003b). Thus, it is possible that Bamman et al.(2001) were measuring increases in IGF-IEa and notMGF at this latter time point.

Whilst it is clear that mechanical activity plays a pivotal role in regulating local IGF-I expression inmuscle, it was unclear as to whether there may befurther regulation, provided by other hormones,notably GH. Some insight into this possibility wasgained from the results of a recent longitudinalstudy where the relationship between exogenousGH administration and strength training exercisewas studied in older people. The subjects (age 74 ±1 year) were assigned to either resistance training(consisting three different lower body exercises: leg press, seated knee extension and seated kneeflexion, which were performed three times a weekand consisted three to five sets of 8–12-RM per session) with placebo, resistance training combinedwith rGH administration alone. GH administrationwithout training did not change MGF mRNA levelswhen measured at 5 weeks (Fig. 14.2), but signific-antly increased IGF-IEa levels (237%). In contrast, 5weeks of resistance training significantly increasedexpression of MGF (163%) and to a lesser extentIGF-IEa (68%). However, when GH treatment wascombined with exercise MGF levels were dramat-ically increased (456%). These data suggest thatexogenous GH administration causes an overall up-regulation of transcription of the IGF-I gene prior tosplicing, which results in more of the primary tran-script of IGF-I. In the absence of strength trainingexercise, splicing is towards the IGF-IEa isoform,but when combined with the mechanical loading it splices towards the MGF isoform. Furthermore, in this study another splice variant, IGF-IEb wasdetected and cloned. The function of this third mus-cle isoform is not yet known.

Page 200: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

184 chapter 14

The structure of IGF-I

IGF-I peptides exist as single chain polypeptidesconsisting of about 70 amino acid residues. Not onlyare these derived by alternative splicing of the IGF-Igene but, like insulin, the initial peptide undergoesa process of post-translational alteration. The prim-ary structure of IGF-I is similar to that of proinsulin(43% sequence homology) as it comprises an aminoterminal B region and an A region that is separatedby a short connecting C domain. Unlike proinsulin,however, it also has a D region extension peptideand an E peptide at its carboxyl terminus (Lowe,1988), and is therefore longer than insulin.

The tertiary structure of IGF-I (Fig. 14.3) was initially predicted using computer graphics. Thiswas based on the three-dimensional crystallinestructure of insulin as determined by X-ray diffrac-tion. As mentioned, IGF-I is somewhat longer thaninsulin although its receptor domain is very similarand acts as a marker for interactive moleculargraphics for visualizing the conformation of IGF-Iand IGF-II peptides. In predicting the tertiary struc-ture of IGF-I, the conservation of the cysteine andglycine residues between IGF-I and proinsulin areparticularly important. The hydrophobic core ofinsulin: A2 Ile, A16 Leu, B12 Val, B15 Leu and B24

Phe (insulin notation) is conserved as well. The mostobvious differences between IGF-I and insulin are inthe C domain. The extra carboxy terminal regionsendow the IGF-I splice variant with special proper-ties which determine their modus operandi. As wellas a hydrophobic core similar to that of proinsulin,IGF-I has three S–S bridges which determine thethree dimensional conformation of this polypep-tide. However the presence of the disulphide linksmake it difficult to synthesise IGF-I chemically andnative function structure and stability require thepresence of all three S–S bridges (Narhi et al. 1993).

Receptors mediate the cellular effects of IGF-I

The biological activity of any hormone depends onthe ability of the target cells to respond to the signalin the extracellular milieu. This is a function of thecell receptors as well as post-receptor mechanisms.IGF-I and IGF-II molecules interact with an array of cell surface receptors that may be present indi-vidually or in various combinations on target cells.Both IGF-I and IGF-II are believed to interact withthe IGF-I receptor (IGF-IR), that is structurally andfunctionally related to the insulin receptor (IR) withwhich it shares > 50% amino acid identity. Despite

% c

han

ge

600

500

400

300

200

100

0

**

#*

*

*MGF5 weeks12 weeks

RT(a) GH only RT + GH

% c

han

ge

350

300

250

200

150

100

50

0

*

*

*

*

*

IGF-IEa5 weeks12 weeks

RT(b) GH only RT + GH

Fig. 14.2 Changes in the expression of mechano growth factor (MGF) (a) and IGF-IEa (b) mRNA after 5 (open bars) and 12 (lined bars) weeks in elderly men in the three intervention groups: Growth hormone only (GH), resistance training only (RT) and resistance training in combination with growth hormone (RT + GH). Values are expressed as a percentagechange from baseline at 5 and 12 weeks. Bars and error bars represent mean values and SEM, respectively. *, Significantdifference from baseline (#) from 5 weeks (P < 0.05). †, Significant difference in % change between isoforms (P < 0.05).(Data taken from Hameed et al. 2004.)

Page 201: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 185

this similarity, IGF-I does not tend to bind with theIR, except at pharmacological doses. This is largelydue to its affinity being approximately two orders ofmagnitude higher for the IGF-IR compared to theIR. As well as binding to the IGF-IR, IGF-II can alsobind to a second receptor known as the IGF-IIR withhigh affinity.

The IGF-IR is a tyrosine specific protein kinasewith an extracellular ligand-binding site. Thisreceptor seems to mediate most actions of IGFs inskeletal muscle. For example, the IGF-IR appears tomediate amino acid and hexose uptake in rat soleusmuscles (Yu & Czech 1984), BC3H1 muscle cells (De Vroede et al. 1984) and DNA synthesis in chickmuscle satellite cells (Duclos et al. 1991). The IGF-Ireceptor is therefore believed to mediate severalactions of IGF-I, such as stimulation of amino aciduptake, proliferation, differentiation and inhibitionof protein degradation (Ewton et al. 1987).

The complexity of IGF-I signaling is increased bythe formation of hybrid receptors that result fromthe dimerization of IGF-IR and IR hemireceptors.Each hybrid receptor consists of a single α and βsubunit linked by disulphide bonds. In some cir-cumstances, these hybrid receptors may outnumber

homoreceptor molecules at the cell surface (reviewedby Le Roith & Roberts 2003). These IGF-IR/IR hybridreceptors bind IGF-I with high affinity, but have areduced affinity for insulin. This can be attributed to the ability IGF-I to bind to either IGF-IR α sub-unit, whereas for insulin to bind effectively requiresinteraction with both the β subunits found in the IR.

It is interesting to note that among known musclegrowth factors, IGF-I is unique in its ability to stimu-late both proliferation and terminal differentiationof post-mitotic myotubes. This may be due to dif-ferent IGF-I splice variants within muscle tissue. Arecent study showed that different IGF-I splice vari-ants have different roles in myoblast proliferationand differentiation (Yang & Goldspink 2002). It alsoshowed that different actions of these differentsplice variants are mediated through a differentreceptor.

IGF-I binding proteins

The roles of the specific binding proteins in deter-mining the autocrine or paracrine actions of the IGFsystem are becoming increasingly apparent (Floriniet al. 1996; Damon et al. 1997). Amongst the seven

5 6

4

78

3

2 1

2928

27

26

25

247

65

4

3

2 1

823

21

22

21

20

19B

D

CA

B

18 16

1715

14

1312

1119 18

910

8

7

6

S-S

S-S

S-S

5

43

2

1

13 14

12

11

10

98

5

67

4

3

2

1

121110

9

17

16

15

20

Fig. 14.3 Tertiary structure ofinsulin-like growth factor I (IGF-I)showing the A, B, C and D domains.Amino acid residues are numberedwithin each domain. The amino Band A domains are separated by ashort interconnecting C domain. TheD domain and E domain (not shown)form the carboxyl terminus of theprotein. As well as the hydrophobiccore similar to that of proinsulin themolecule has three disulfide bridges(shown as S–S), which determine itsthree-dimensional conformation.Two of these are located in the core of the molecule (B18–A20; A6–A11)and the third on the surface (B6–A7).(Adapted from Blundell et al. 1983.)

Page 202: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

186 chapter 14

IGFBPs described so far, four (IGFBP-2, 4, 5 and 6)are produced by different myoblast cell lineswhereas only IGFBP-4, 5 and 6 are expressed byadult skeletal muscle (Florini et al. 1996; Putzer et al.1998). IGFBPs have been associated with the sys-temic IGF system, but their expression in skeletalmuscle would have the effect of retaining IGF-Iwithin the muscle tissue. In the unbound state theIGF-I peptides have a short half-life and the IGFBPswere previously thought of as carrier proteins in theserum. However, when expressed within the mus-cle they modulate the endocrine as well as the localinfluences of IGF-I (Mohan et al. 1996). The preciseactions of IGFBPs at the tissue level are unknown,but it is apparent that they stabilize and augmentlocal IGF-I bioavailability (Jones & Clemmons 1995;Mohan et al. 1996; Clemmons et al. 1998).

Both mRNA and protein levels of IGF-I andIGFBPs have been shown to be up-regulated duringregeneration after ischaemic injury (Jennische &Hall 2000). In situ hybridization studies have shownIGFBP-5 to be restricted to regenerating musclecells, whereas connective tissue cells expressedIGFBP-4 (Boes et al. 1992). Mechanically loading, orunloading the muscle, has been shown to regulateseveral of the binding proteins in muscle. For ex-ample, Awede et al. (1999) reported that overload-ing the muscles in mice increased the expression of IGFBP-4 mRNA, but decreased that of IGFBP-5.In contrast, unloading of mouse muscle resulted inreduced levels of IGFBP5 mRNA, but did not affectlevels of IGFBP-4. Both of these binding proteinswere assumed to mediate the effects of IGF-I via re-gulation of the free IGF-I concentration in muscleand possibly via competition with IGF receptors forIGF-I (Awede et al. 1999). Experiments are under-way to characterize the specific binding protein forMGF, which differs from the other binding proteinsfor the other splice variants.

Biological action of the IGF-I splicevariants

All the IGF-I splice variants have the same receptor-binding domain encoded by exons 3 and 4. This isapparently responsible for the anabolic effects ofIGF-I. These have been clearly demonstrated by

numerous in vitro studies, where it has been shownthat IGF-I acts to increase the diameter of myotubes,suppress protein degradation, increase amino aciduptake and stimulate protein synthesis (Ewton et al.1987; Vandenburgh et al. 1991; Florini et al. 1996;Semsarian et al. 1999; Bodine et al. 2001; Rommel et al. 2001). Its expression during muscle hyper-trophy has been shown by using several animalmodels, including stretch-induced hypertrophy ofthe muscle. For example, Schlechter et al. (1986) andCzerwinski et al. (1994) reported an increasedexpression of muscle IGF-I mRNA, and DeVol et al.(1990) demonstrated that there was a threefoldincrease in IGF-I mRNA levels in the soleus andplantaris muscles in 11–12-week-old female rats fol-lowing tenotomy-induced hypertrophy. This par-ticular study employed hypophysectomized rats,which further suggests that the observed increase inIGF-I mRNA expression was GH independent.Later studies utilizing a similar model of functionaloverload in both normal and hypophysectomizedrats found that both mRNA and protein levels ofIGF-I were increased in muscle, prior to the attain-ment of significant hypertrophy, and remained elev-ated for up to 28 days during the hypertrophy process (Adams & Haddad 1996). In another studywhich utilized treadmill training of GH-suppressedrats, levels of IGF-I mRNA and protein increased by55% and 250% respectively (Zanconato et al. 1994).Furthermore, overexpression (Coleman et al. 1995)or direct infusion (Adams & McCue 1998) of IGF-I inmuscle results in hypertrophy, whereas inhibitionof intracellular signaling components associatedwith IGF-IR activation can prevent this response(Bodine et al. 2001). Another study, examining theassociation between local IGF-I overexpression andatrophy induced by hind limb unloading, con-cluded that overexpression of IGF-I in the musclesof transgenic mice was not shown to preventunloading-induced atrophy (Criswell et al. 1998).

Genetic manipulation of IGF-I in muscle

Transgenically modified mice that overexpress theIGF-I gene have been produced in several laborator-ies. Initial experiments of this kind by Mathews et al.(1988) in which the GH gene was overexpressed,

Page 203: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 187

reported a 30% increase in muscle and an increase inbone density (Mathews et al. 1988). These experi-ments involved using a metallothionein promoter,which when activated increased the expression ofIGF-I in most tissues. Later, experiments involvedIGF-I transgenes that were under the control ofmuscle regulatory elements, such as the α-actin pro-moter (Coleman et al. 1995). Although serum levelsof IGF-I were not significantly elevated in thesemice, there was a considerable increase in musclemass. More recently, work by the same group hasshown that both muscle and motor neuron regen-eration was enhanced in these mice when comparedto wild type (Rabinovsky et al. 2003). It is difficult todetermine whether the full length cDNA has beenused in these gene transfer experiments and if thisinsert includes the sequence for a binding protein.This is important if we are to understand whether or not the expression of IGF-I is muscle specific; thisis usually determined by whether the introducedDNA is under the control of a muscle specific regu-latory sequence. IGF-I knock-out mice (lacking theentire IGF-I gene sequence) have also been pro-duced. Therefore, none of the IGF-I peptide can betranscribed (Baker et al. 1993). However, these micedo not survive very long after birth, as their musclesappear to be dystrophic. Interestingly, the results of recent tissue specific gene deletion experimentsusing the Cre-loxP model of gene deletion havequestioned the role of GH and liver IGF-I in control-ling postnatal growth and development (Sjogren et al. 1999; Yakar et al. 1999). This homologousrecombination system was used to create a liverspecific deletion of the IGF-I gene but allowed nor-mal expression of this gene in other non-hepatic tissues, such as heart, muscle, fat, spleen and kid-ney. The effect of this liver specific gene deletion of IGF-I on growth and development in these micewas to reduce circulating IGF-I levels at 6 weeks ofage compared with wild type animals. Interestingly,measurements of body size and individual organweights at 6 weeks showed no difference betweenthe ‘knockout’ animals and their wild type litter-mates. Thus, postnatal growth and developmentproceeded without the contribution of liver derivedIGF-I (Sjogren et al. 1999), emphasizing the role ofthe local IGF-I system in the process.

Gene transfer of the different IGF-Isplice variants

The finding that muscle fibers can be transfected bya simple intramuscular injection of a plasmid vectorcontaining the cDNA sequence of a gene of interestprovides a method for treating medical conditionsthat are associated with marked muscle loss. Morerecently, experiments in which constructs con-taining the IGF-I splice variants, including MGFhave been performed. For example, in one suchexperiment a plasmid construct containing MGFcDNA was injected into the muscles of normal miceto determine the role of the MGF splice variant inmuscle maintenance. This study resulted in a 25%increase in the mean muscle fiber size in injectedmuscle within 2 weeks, and this was shown to bedue to an increase in the size of the muscle fibers(Goldspink & Yang 2001). Similar experiments byother groups have also been carried out using a viralconstruct containing the liver type of IGF-I (IGF-IEa). This also resulted in a less than 20% increase inmuscle mass, but this took over 4 months to develop(Barton-Davis et al. 1998). As well as treating certainmedical conditions, gene transfer of this type maywell be open to abuse. The use of an adenoviral vector for delivery would make detection relativelysimple using PCR based methods, as this virusinfects most cell types. Plasmid vectors are in con-trast more difficult to detect, but new approachesthat involve determining the different actions of thegene products from those of introduced gene areunderway.

IGF-I signaling pathways in musclehypertrophy

The signaling pathways by which IGF-I promotesskeletal muscle hypertrophy remain unclear, withroles suggested for both the calcineurin/NFAT(nuclear factor of activated T-cells) pathway (Musaroet al. 1999; Semsarian et al. 1999) and the P13-kinase/Akt pathway (Rommel et al. 2001). More recently,studies investigating the hypertrophic responseboth in vitro and in vivo have reported that it is theAkt/mTOR pathway and not the calcineurin path-way which is involved in promoting hypertrophy,

Page 204: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

188 chapter 14

by activating downstream targets such as P > 0.056kinase (Bodine et al. 2001; Rommel et al. 2001). Inaddition, it was also reported that IGF-I might infact act via Akt to inhibit the calcineurin/NFAT sig-naling pathway during this process (Rommel et al.2001). Unfortunately studies conducted to date havelooked at the expression of total IGF-I in response tomuscle overload.

IGF-I and sarcopenia

Increasing age is associated with a loss of muscleand mass and function, but the mechanisms under-lying this process remain unclear. There is a reduc-tion in circulating levels of GH and IGF-I (Rudmanet al. 1981) and an actual loss of muscle fibers (Lexellet al. 1988). There is some emerging evidence thatIGF-I has a role to play the maintenance of muscle in later life. Using a recombinant adeno-associatedvirus containing a myosin light chain (MLC1/3)promoter, Barton-Davis et al. (1998) injected thecDNA of an IGF-I construct into the extensor digi-torum longus (EDL) muscles of young (6 months) and old (27 months) mice. This resulted in overex-pression of IGF-I (Ea isoform) in this muscle, but didnot give rise to elevated circulating levels of IGF-I in plasma. Four months post-injection, the injectedmuscle of the younger animals was on average 15%larger and stronger than the non-injected muscle. In the older animals, the injected muscle was 27%stronger when compared with the non-injected oldanimals, resulting in similar values for mass andfunction to the 6-month-old animals. An age-relatedimprovement in muscle mass and function was alsodemonstrated in a very recent study using a mousemodel in which transgenic animals were bred tooverexpress the IGF-IEa gene (which they termedm.IGF-I) (Musaro et al. 2001). At 6 months of age, thefibers of the transgenic animals were 32 µm in dia-meter as compared to 18 µm in the wild type animals.There was, however, preferential hypertrophy ofthe faster fiber types (46 µm in the transgenic animalvs. 32 µm in the wild type) with little effect on theslow fibers (16 µm vs. 18 µm). This was attributed tothe low expression of the MLC regulatory cassette(1/3 locus) in slow muscle fibers. In the older trans-genic animals, muscle mass was maintained in

animals 20 months of age, whilst it was significantlyatrophied in the wild type animals. These geneticmanipulation studies suggest that local IGF-I maybe an important factor in maintaining muscle massin old age. Recently, Owino et al. (2001) used tendonablation to overload the soleus and plantaris mus-cles of rats to study any age-related differences inthe response of the muscle IGF-I isoforms. Animalsof different ages (4, 12, 24 months) were overloadedfor 5 days. They reported that the MGF mRNA wasup-regulated by ~ 1200% in the young animalswhen compared to the control leg, but was up-regu-lated to a significantly less extent (~ 500%) in the old animals. The Ea isoform was also up-regulated, butshowed no clear age-related effect. In recent studieson humans, and as mentioned previously, Hameedet al. (2003a) reported that older men showed areduced MGF response when measured shortlyafter a single bout of knee extensor weightliftingexercise compared to young subjects. However,Singh et al. (1999) showed that overall muscle IGF-Ilevels could be increased in older people as a resultof a 10-week period (three times per week) ofstrength training exercise. As discussed previously,Hameed et al. (2004) showed that such a trainingregimen could cause an increase in the level of tran-script expression all three of the IGF-I splice variants(IGF-IEa, IGF-IEb and MGF) in the muscles of olderpeople. Interestingly, it was MGF that showed themost marked increase (see Figure 14.2). The abilityto do this no doubt contributes to the retained capa-city, of even very elderly muscle, to hypertrophy inresponse to strength training exercise.

Mechano growth factor, satellite cellsand muscle damage

Satellite cells in skeletal muscle were first describedby Mauro (1961). It is now realized that these cellsprovide the extra nuclei for postnatal growth (Moss& Leblond 1970; Schultz 1996) and that they are alsoinvolved in repair and regeneration following localinjury of muscle fibers (Grounds 1998). In normaladult undamaged tissue, the satellite cells are quiescent and usually detected just beneath thebasal lamina. They express M-cadherin (M-cad)(Bornemann & Schmalbruch 1994; Irintchev et al.

Page 205: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 189

1994) when activated, and commence to coexpressmyogenic factors including c-met, MyoD and myf5and later myogenin (Cornelison & Wold 1997;Beauchamp et al. 2000; Qu-Petersen et al. 2002). Theorigin of satellite cells is still somewhat uncertain, as they were thought to be residual myoblasts(reviewed by Seale & Rudnicki 2000), but there isaccumulating evidence that some may also origin-ate from pluripotent stem cells derived from pro-genitor cells of the vasculature (Qu-Petersen et al.2002). Pluripotent stem cells from bone marrowcells (Ferrari et al. 1998), as well as epidermal cells(Pye & Watt 2001), have also been shown to fuse andadopt the muscle phenotype when introduced into dystrophic muscle.

It has been established that even in normal mus-cle, local injury does occur from time to time(Wernig et al. 1990), but in certain diseases such asthe muscular dystrophies, the muscle fibers aremarkedly more susceptible to damage, in particularto the membrane (Cohn & Campbell 2000). The contractile system of muscle fibers also sustainsdamage during eccentric contractions, that is to saywhen the muscle is activated whilst being stretched.It is interesting to note that the forces generated byactivation combined with stretch exceed even thoseof a maximal isometric contraction. In the musclefibers involved, the sarcomeres may be pulled out to such a degree that there is no longer any overlapof the actin and myosin filaments, thus causingdamage (Lieber & Friden 1999).

During regeneration of skeletal muscle in youngrats following ischaemia or myotoxin-induced dam-age, elevated expression of IGF-I has been reported(Jennische & Hansson 1987; Jennische et al. 1987;Edwall et al. 1989) which was diminished by the15th day of recovery (Marsh et al. 1997). In a morerecent study, the response of the different isoformsof IGF-I, IGF-IEa and MGF to such stimuli werestudied for the first time and related to the activa-tion of muscle satellite (stem) cells in vivo (Hill & Goldspink 2003). Results of the experiments, in which damage was induced by bupivacaine,demonstrated a surge of IGF-IEa mRNA expressionthat was maximal at 11 days and diminished there-after to similar levels as those in the non-injectedanimals. On the other hand, MGF mRNA showed a

much earlier transient response, which peaked at 4 days post-bupivacaine injection and decreasedthereafter. Following mechanical damage, the peakin MGF expression occurred even earlier. It seemsthat in both myotoxin- and mechanical activity-induced damage models, the temporal expressionpattern for each IGF-I splice variant showed sim-ilar differential gene splicing sequences, with MGFpeaking before IGF-IEa. This temporal difference inexpression of the two muscle IGF-I mRNA tran-scripts has also been described in the rat followingcommencement of resistance exercise (Haddad &Adams 2002). As M-cad expression peaked wellbefore IGF-IEa, whether it was measured as mRNAor protein, it is unlikely that the systemic type of IGF-IEa is responsible for initial activation ofsatellite cells. However, it is not possible to say fromthese data whether this was due to an increase innumber of satellite cells as it is known that quiescentsatellite cells do stain to some extent for M-cad pro-tein (Rosenblatt et al. 1994). Nevertheless, it repres-ents a marked increase of M-cad whether it is inexisting satellite cells or an increase in the number ofthese cells or both.

MGF and IGF-IEa splice variants apparently yieldthe same mature peptide, which is derived from thehighly conserved exons 3 and 4 of the IGF-I gene.These exons, present in all known IGF splice vari-ants are known to encode the IGF-I receptor liganddomain. A mechanism of extracellular endoproteo-lysis of the IGF-I prohormone results in the samemature peptide (Gilmour 1994), even though thesplice variants of IGF-I may have different 3’sequences including the E domain. It has been sug-gested that IGF-I precursors could be pluripotent, ina form analogous to that of the prohormone pro-piomelanocortin and proglucagon (Siegfried et al.1992). The observation that a synthetic peptidederived from the rat Eb domain induces prolifera-tion in epithelial cells is noteworthy (Siegfried et al.1992). The growth promoting properties of the MGFE peptide and its role as an independent growth fac-tor is supported by recent cell culture experimentswhere stable transfection with MGF was shown tostimulate myoblast proliferation but differentiationwas suppressed. The addition of a synthetic MGFpeptide or the medium from MGF-transfected cells

Page 206: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

190 chapter 14

onto normal C2C12 cells also inhibited their differ-entiation. Yet this inhibition was reversed when thepeptide or the medium were withdrawn. In con-trast, cells of the liver type, systemic IGF-I (IGF-IEa)positive clone did form myotubes and the normalcell lines showed less cellular proliferation as wellas forming myotubes. Of particular interest was theobservation that when an IGF-I receptor antibodywas added to the muscle cell cultures, cell prolifera-tion induced by MGF was not inhibited, whereastheir stimulation to increase in mass and to formmyotubes by IGF-I was reduced. These data stronglysuggest that MGF is involved in another signalingpathway in addition to that associated with the IGF-I receptor (Yang & Goldspink 2002).

The results of these studies provide additionalinsight into the complexity and implication of theIGF-I system in conditions of damage and sub-sequent regeneration. IGF-IEa and MGF are pro-duced by the active muscle in rodents and humansand have been shown to be positive regulators ofmuscle hypertrophy (McKoy et al. 1999; Goldspink2001; Owino et al. 2001; Hameed et al. 2003). How-ever, as reported here, the MGF isoform is acutelyinduced, whereas IGF-IEa has a delayed effect thatis sustained during the later phase of regeneration.When comparing mechanical damage with myotoxindamage it is apparent that both involve a relativelyrapid expression of the MGF splice variant, althoughit may seem that this growth/repair factor has beenmisnamed ‘mechano growth factor’. However, evenin the case of myotoxin-induced damage it is likelythat the damaged tissue mass is subjected toincreased mechanical strain. Also, it is known thatthe cells swell following damage, thus resulting inthe same cellular response. As the expression of theautocrine splice variant (MGF) precedes satellite cellactivation it is likely that this form of IGF-I is asso-ciated with satellite cell activation, not the systemicIGF-IEa type. This is in accord with the finding

that MGF is not appropriately expressed in dys-trophic muscles (Goldspink 1996) and the decreasein MGF mRNA levels in response to mechanicaloverload in older muscles (Owino et al. 2001). Thereis a deficiency of active satellite cells in both thesesituations in which local tissue repair becomesincreasingly impaired. Future experiments invest-igating the expression of the two transcripts and activation of satellite cells in young and old musclesafter the therapeutic application of MGF and IGF-IEa to ameliorate muscle loss are in progress.

Summary

It is becoming increasingly clear that muscle adapta-tion to high resistance exercise as well as its res-ponse to contraction induced damage or otherwise,is mediated at the local level by local growth andrepair factors. The IGF-I family comprises membersthat play different roles in the growth and repairprocesses. Alternative splicing is a subtle andsophisticated mechanism by which different IGF-Iisoforms can be generated from the same gene tohave different biological roles. Understanding theseroles and how these IGF-I isoforms may interactwith other control processes in muscle (e.g. testos-terone, myostatin, ubiquitin, etc.) will help in optimizing training regimens for athletes. Further-more, an understanding of the signaling processestriggered by exercise training will also provide uswith the means to develop more effective methodsfor testing exogenous enhancing substances used indoping.

Acknowledgments

This work was supported by grants from theWellcome Trust, the World Anti-Doping Agency(WADA) and an EC (PENAM) grant for studyingthe effects of exercise including muscle damage.

References

Adams, G.R. (2002) Invited review:autocrine/paracrine IGF-I and skeletalmuscle adaptation. Journal of AppliedPhysiology 93, 1159–1167.

Adams, G.R. & Haddad, F. (1996) The

relationships among IGF-1, DNAcontent, and protein accumulationduring skeletal muscle hypertrophy.Journal of Applied Physiology 81,2509–2516.

Adams, G.R. & McCue, S.A. (1998)Localized infusion of IGF-I results inskeletal muscle hypertrophy in rats.Journal of Applied Physiology 84,1716–1722.

Page 207: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 191

Awede, B., Thissen, J., Gailly, P. & Lebacq,J. (1999) Regulation of IGF-I, IGFBP-4and IGFBP-5 gene expression by loadingin mouse skeletal muscle. FEBS Letters461, 263–267.

Baker, J., Liu, J.P., Robertson, E.J. &Efstratiadis, A. (1993) Role of insulin-like growth factors in embryonic andpostnatal growth. Cell 75, 73–82.

Bamman, M.M., Shipp, J.R., Jiang, J. et al.(2001) Mechanical load increases muscleIGF-I and androgen receptor mRNAconcentrations in humans. AmericanJournal of Physiology. Endocrinology andMetabolism 280, E383–E390.

Barton-Davis, E.R., Shoturma, D.I.,Musaro, A., Rosenthal, N. & Sweeney,H.L. (1998) Viral mediated expression of insulin-like growth factor I blocks theaging-related loss of skeletal musclefunction. Proceedings of the NationalAcademy of Sciences of the United States of America 95, 15 603–15 607.

Baumann, G. (1991) Growth hormoneheterogeneity: genes, isohormones,variants, and binding proteins. EndocrineReviews 12(4), 424–449.

Beauchamp, J.R., Heslop, L., Yu, D.S. et al.(2000) Expression of CD34 and Myf5defines the majority of quiescent adultskeletal muscle satellite cells. Journal ofCell Biology 151, 1221–1234.

Beshyah, S.A., Freemantle, C., Thomas, E.& Johnston, D.G. (1995) Comparison ofmeasurements of body composition bytotal body potassium, bioimpedanceanalysis, and dual-energy X-rayabsorptiometry in hypopituitary adultsbefore and during growth hormonetreatment. American Journal of ClinicalNutrition 61, 1186–1194.

Blundell, T.L., Bedarkar, S. & Humbel, R.E.(1983) Tertiary structures, receptorbinding and antigenicity of insulin likegrowth factors. Federation Proceedings 42,2592–2597.

Bodine, S.C., Stitt, T.N., Gonzalez, M. et al.(2001) Akt/mTOR pathway is a crucialregulator of skeletal muscle hypertrophyand can prevent muscle atrophy in vivo.Nature Cell Biology 3, 1014–1019.

Boes, M., Booth, B.A., Sandra, A. et al.(1992) Insulin-like growth factor bindingprotein (IGFBP)4 accounts for theconnective tissue distribution ofendothelial cell IGFBPs perfusedthrough the isolated heart. Endocrinology131, 327–330.

Bornemann, A. & Schmalbruch, H. (1994)Immunocytochemistry of M-cadherin inmature and regenerating rat muscle.Anatomical Records 239, 119–125.

DeVol, D.L., Rotwein, P., Sadow, J.L.,Novakofski, J. & Bechtel, P.J. (1990)Activation of insulin-like growth factorgene expression during work-inducedskeletal muscle growth. American Journalof Physiology 259, E89–E95.

De Vroede, M.A., Romanus, J.A.,Standaert, M.L. et al. (1984) Interaction of insulin-like growth factors with anonfusing mouse muscle cell line:binding, action, and receptor down-regulation. Endocrinology 114, 1917–1929.

Duclos, M.J., Wilkie, R.S. & Goddard, C.(1991) Stimulation of DNA synthesis inchicken muscle satellite cells by insulinand insulin-like growth factors:evidence for exclusive mediation by a type-I insulin-like growth factorreceptor. Journal of Endocrinology 128,35–42.

Edwall, D., Schalling, M., Jennische, E. & Norstedt, G. (1989) Induction ofinsulin-like growth factor I messengerribonucleic acid during regeneration ofrat skeletal muscle. Endocrinology 124,820–825.

Ewton, D.Z., Falen, S.L. & Florini, J.R.(1987) The type II insulin-like growthfactor (IGF) receptor has low affinity forIGF-I analogs: pleiotypic actions of IGFson myoblasts are apparently mediatedby the type I receptor. Endocrinology 120,115–123.

Ferrari, G., Cusella-De Angelis, G., Coletta,M. et al. (1998) Muscle regeneration by bone marrow-derived myogenicprogenitors. Science 279, 1528–1530.

Florini, J.R., Ewton, D.Z. & Coolican, S.A.(1996) Growth hormone and the insulin-like growth factor system in myogenesis.Endocrine Reviews 17, 481–517.

Gilmour, R.S. (1994) The implications of insulin-like growth factor mRNAheterogeneity. Journal of Endocrinology140, 1–3.

Goldspink, G. (1996) Muscle growth andmuscle function: a molecular biologicalperspective. Research in VeterinaryScience 60, 193–204.

Goldspink, G. (2001) Method of treatingmuscular disorders. United States PatentNo: US 6 221 842 B1.

Goldspink, G. & Yang, S.Y. (2001) Methodof treating muscular disorders. UnitedStates Patent No: 6 221 842 B1.

Goldspink, G., Scutt, A., Loughna, P.T. et al. (1992) Gene expression in skeletalmuscle in response to stretch and forcegeneration. American Journal ofPhysiology 262, R356–R363.

Green, H., Morikawa, M. & Nixon, T.(1985) A dual effector theory of

Chew, S.L., Lavender, P., Clark, A.J. &Ross, R.J. (1995) An alternatively splicedhuman insulin-like growth factor-Itranscript with hepatic tissue expressionthat diverts away from the mitogenicIBE1 peptide. Endocrinology 136,1939–1944.

Clemmons, D.R., Busby, W., Clarke, J.B. et al. (1998) Modifications of insulin-likegrowth factor binding proteins and theirrole in controlling IGF actions. EndocrineJournal 45 (suppl.), S1–S8.

Cohn, R.D. & Campbell, K.P. (2000)Molecular basis of musculardystrophies. Muscle and Nerve 23,1456–1471.

Coleman, M.E., DeMayo, F., Yin, K.C. et al.(1995) Myogenic vector expression ofinsulin-like growth factor I stimulatesmuscle cell differentiation and myofiberhypertrophy in transgenic mice. Journal of Biological Chemistry 270, 12109–12 116.

Cornelison, D.D. & Wold, B.J. (1997)Single-cell analysis of regulatory geneexpression in quiescent and activatedmouse skeletal muscle satellite cells.Developmental Biology 191, 270–283.

Criswell, D.S., Booth, F.W., DeMayo, F. et al. (1998) Overexpression of IGF-I inskeletal muscle of transgenic mice doesnot prevent unloading-induced atrophy.American Journal of Physiology 275,E373–E379.

Cuneo, R.C., Salomon, F., Wiles, C.M.,Hesp, R. & Sonksen, P.H. (1991) Growthhormone treatment in growth hormone-deficient adults. I. Effects on musclemass and strength. Journal of AppliedPhysiology 70, 688–694.

Czerwinski, S.M., Martin, J.M. & Bechtel,P.J. (1994) Modulation of IGF mRNAabundance during stretch-inducedskeletal muscle hypertrophy andregression. Journal of Applied Physiology76, 2026–2030.

Damon, S.E., Haugk, K.L., Swisshelm, K. & Quinn, L.S. (1997) Developmentalregulation of Mac25/insulin-like growthfactor-binding protein-7 expression inskeletal myogenesis. Experimental CellResearch 237, 192–195.

Daughaday, W.H. & Reeder, C. (1966)Synchronous activation of DNAsynthesis in hypophysectomized ratcartilage by growth hormone. Journal of Laboratory and Clinical Medicine 68,357–368.

Daughaday, W.H., Hall, K., Raben, M.S. et al. (1972) Somatomedin: proposeddesignation for sulphation factor. Nature235, 107.

Page 208: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

192 chapter 14

growth-hormone action. Differentiation29, 195–198.

Grounds, M.D. (1998) Age-associatedchanges in the response of skeletalmuscle cells to exercise andregeneration. Annals of the New YorkAcademy of Sciences 20(854), 78–91.

Haddad, F. & Adams, G.R. (2002) Selectedcontribution: acute cellular andmolecular responses to resistanceexercise. Journal of Applied Physiology 93,394–403.

Hameed, M., Orrell, R.W., Cobbold, M.,Goldspink, G. & Harridge, S.D. (2003a)Expression of IGF-I splice variants inyoung and old human skeletal muscleafter high resistance exercise. Journal ofPhysiology 547, 247–254.

Hameed, M., Toft, A.D., Pedersen, B.K.,Harridge, S.D. & Goldspink, G. (2003b)The effects of eccentric exercise on IGF-Iisoform expression in the muscle ofyoung and elderly people. Journal ofMuscle Research and Cell Motility 24,323–386.

Hameed, M., Lange, K.H., Andersen, H.,Schjerling, P., Kjær, M., Harridge, S.D.R.& Goldspink, G. et al. (2004) The effect ofrecombinant human growth hormoneand resistance training on IGF-I mRNAexpression in the muscles of elderlymen. Journal of Physiology 15(1), 231–240.

Hellsten, Y., Hansson, H.A., Johnson, L.,Frandsen, U. & Sjodin, B. (1996)Increased expression of xanthineoxidase and insulin-like growth factor I(IGF-I) immunoreactivity in skeletalmuscle after strenuous exercise inhumans. Acta Physiologica Scandinavica157, 191–197.

Hill, M. & Goldspink, G. (2003) Expressionand splicing of the insulin-like growthfactor gene in rodent muscle isassociated with muscle satellite (stem)cell activation following local tissuedamage. Journal of Physiology 549,409–418.

Irintchev, A., Zeschnigk, M., Starzinski-Powitz, A. & Wernig, A. (1994)Expression pattern of M-cadherin innormal, denervated, and regeneratingmouse muscles. Developmental Dynamics199, 326–337.

Jennische, E. & Hall, C.M. (2000)Expression and localisation of IGF-binding protein mRNAs in regeneratingrat skeletal muscle. APMIS: ActaPathologica, Microbiologica, etImmunologica Scandinavica 108(11),747–755.

Jennische, E. & Hansson, H.A. (1987)Regenerating skeletal muscle cells

Mathews, L.S., Hammer, R.E., Behringer,R.R. et al. (1988) Growth enhancement of transgenic mice expressing humaninsulin-like growth factor I.Endocrinology 123, 2827–2833.

Mauro, A. (1961) Satellite cell of skeletalmuscle fibers. Journal of Biophysical andBiochemical Cytology 9, 493–495.

Mohan, S., Baylink, D.J., Pettis, J.L. (1996)Insulin-like growth factor (IGF)-bindingproteins in serum––do they haveadditional roles besides modulating theendocrine IGF actions? Journal of ClinicalEndocrinology and Metabolism 81(11),3817–20.

Moss, F.P. & Leblond, C.P. (1970) Nature of dividing nuclei in skeletal muscle ofgrowing rats. Journal of Cell Biology 44,459–462.

Musaro, A., McCullagh, K.J., Naya, F.J.,Olson, E.N. & Rosenthal, N. (1999) IGF-1induces skeletal myocyte hypertrophythrough calcineurin in association withGATA-2 and NF-ATc1. Nature 400,581–585.

Musaro, A., McCullagh, K.J., Paul, A. et al. (2001) Localized IGF-1 transgeneexpression sustains hypertrophy andregeneration in senescent skeletalmuscle. Nature Genetics 27, 195–200.

Narhi, L.O., Hua, Q.X., Arakawa, T. et al.(1993) Role of native disulfide bonds inthe structure and activity of insulin-likegrowth factor 1: genetic models ofprotein-folding intermediates.Biochemistry 32, 5214–5221.

Nindl, B.C., Kraemer, W.J., Marx, J.O.,Tuckow, A.P. & Hymer, W.C. (2003)Growth hormone molecularheterogeneity and exercise. Exercise andSport Sciences Reviews 31(4), 161–166.

Owino, V., Yang, S.Y. & Goldspink, G.(2001) Age-related loss of skeletalmuscle function and the inability toexpress the autocrine form of insulin-like growth factor-1 (MGF) in responseto mechanical overload. FEBS Letters505, 259–263.

Putzer, P., Breuer, P., Gotz, W. et al. (1998) Mouse insulin-like growth factor binding protein-6: expression,purification, characterization andhistochemical localization. Molecular andCellular Endocrinology 137, 69–78.

Pye, D. & Watt, D.J. (2001) Dermalfibroblasts participate in the formationof new muscle fibres when implantedinto regenerating normal mouse muscle.Journal of Anatomy 198, 163–173.

Qu-Petersen, Z., Deasy, B., Jankowski, R. et al. (2002) Identification of a novelpopulation of muscle stem cells in

express insulin-like growth factor I. Acta Physiologica Scandinavica 130,327–332.

Jennische, E., Skottner, A. & Hansson, H.A.(1987) Satellite cells express the trophicfactor IGF-I in regenerating skeletalmuscle. Acta Physiologica Scandinavica129, 9–15.

Jones, J.I. & Clemmons, D.R. (1995) Insulin-like growth factors and their bindingproteins: biological actions. EndocrineReviews 16, 3–34.

Klapper, D.G., Svoboda, M.E. & Van Wyk,J.J. (1983) Sequence analysis ofsomatomedin-C: confirmation ofidentity with insulin-like growth factorI. Endocrinology 112, 2215–2217.

Lange, K.H., Andersen, J.L., Beyer, N. et al. (2002) GH administration changesmyosin heavy chain isoforms in skeletalmuscle but does not augment musclestrength or hypertrophy, either alone or combined with resistance exercisetraining in healthy elderly men. Journalof Clinical Endocrinology and Metabolism87, 513–523.

Le Roith, D. & Roberts, C.T., Jr. (2003) Theinsulin-like growth factor system andcancer. Cancer Letters 195, 127–137.

Lexell, J., Taylor, C.C. & Sjostrom, M.(1988) What is the cause of the ageingatrophy? Total number, size andproportion of different fiber typesstudied in whole vastus lateralis musclefrom 15- to 83-year-old men. Journal ofthe Neurological Sciences 84, 275–294.

Lieber, R.L. & Friden, J. (1999) Mechanismsof muscle injury after eccentriccontraction. Journal of Science andMedicine in Sport/Sports MedicineAustralia 2, 253–265.

Lowe, W.L., Jr., Lasky, S.R., LeRoith, D. & Roberts, C.T., Jr. (1988) Distributionand regulation of rat insulin-like growthfactor I messenger ribonucleic acidsencoding alternative carboxyterminal E-peptides: evidence for differentialprocessing and regulation in liver.Molecular Endocrinology (Baltimore, Md)2, 528–535.

McKoy, G., Ashley, W., Mander, J. et al.(1999) Expression of insulin growthfactor-1 splice variants and structuralgenes in rabbit skeletal muscle inducedby stretch and stimulation. Journal ofPhysiology 516(2), 583–592.

Marsh, D.R., Criswell, D.S., Hamilton,M.T. & Booth, F.W. (1997) Association of insulin-like growth factor mRNAexpressions with muscle regeneration in young, adult, and old rats. AmericanJournal of Physiology 273, R353–R358.

Page 209: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

igf-i splice variants in muscle growth 193

mice: potential for muscle regeneration.Journal of Cell Biology 157, 851–864.

Rabinovsky, E.D., Gelir, E., Gelir, S. et al.(2003) Targeted expression of IGF-1transgene to skeletal muscle acceleratesmuscle and motor neuron regeneration.FASEB Journal 17, 53–55.

Rees, C. & Clemmons, D.R. (1998)Inhibition of IGFBP-5 binding toextracellular matrix and IGF-I-stimulated DNA synthesis by a peptidefragment of IGFBP-5. Journal of CellularBiochemistry 71, 375–381.

Rinderknecht, E. & Humbel, R.E. (1978)The amino acid sequence of humaninsulin-like growth factor I and itsstructural homology with proinsulin.Journal of Biological Chemistry 253,2769–2776.

Roberts, P., McGeachie, J.K. & Grounds,M.D. (1997) The host environmentdetermines strain-specific differences in the timing of skeletal muscleregeneration: cross-transplantationstudies between SJL/J and BALB/cmice. Journal of Anatomy 191(4), 585–594.

Rommel, C., Bodine, S.C., Clarke, B.A. et al. (2001) Mediation of IGF-1-inducedskeletal myotube hypertrophy by PI(3)K/Akt/mTOR andPI(3)K/Akt/GSK3 pathways. NatureCell Biology 3, 1009–1013.

Rosenblatt, J.D., Yong, D. & Parry, D.J.(1994) Satellite cell activity is requiredfor hypertrophy of overloaded adult ratmuscle. Muscle and Nerve 17, 608–613.

Rotwein, P., Pollock, K.M., Didier, D.K. & Krivi, G.G. (1986) Organization andsequence of the human insulin-likegrowth factor I gene. Alternative RNAprocessing produces two insulin-likegrowth factor I precursor peptides.Journal of Biological Chemistry 261,4828–4832.

Rudman, D., Kutner, M.H., Rogers, C.M. et al. (1981) Impaired growth hormonesecretion in the adult population:

Wernig, A., Irintchev, A. & Weisshaupt, P.(1990) Muscle injury, cross-sectionalarea and fibre type distribution in mouse soleus after intermittent wheel-running. Journal of Physiology 428,639–652.

Yakar, S., Liu, J.L., Stannard, B. et al. (1999)Normal growth and development in theabsence of hepatic insulin-like growthfactor I. Proceedings of the NationalAcademy of Sciences of the United States ofAmerica 96, 7324–7329.

Yang, S.Y. & Goldspink, G. (2002) Differentroles of the IGF-I Ec peptide (MGF) andmature IGF-I in myoblast proliferationand differentiation. FEBS Letters 522,156–160.

Yang, S.Y., Alnaqeeb, M., Simpson, H. & Goldspink, G. (1996) Cloning andcharacterization of an IGF-I isoformexpressed in skeletal muscle subjected to stretch. Journal of Muscle Research andCell Motility 17, 487–495.

Yarasheski, K.E., Campbell, J.A., Smith, K.et al. (1992) Effect of growth hormoneand resistance exercise on musclegrowth in young men. American Journalof Physiology 262, E261–E267.

Yarasheski, K.E., Zachwieja, J.J., Campbell,J.A. & Bier, D.M. (1995) Effect of growthhormone and resistance exercise onmuscle growth and strength in oldermen. American Journal of Physiology 268,E268–E276.

Yu, K.T. & Czech, M.P. (1984) The type Iinsulin-like growth factor receptormediates the rapid effects ofmultiplication-stimulating activity onmembrane transport systems in ratsoleus muscle. Journal of BiologicalChemistry 259, 3090–3095.

Zanconato, S., Moromisato, D.Y.,Moromisato, M.Y. et al. (1994) Effect of training and growth hormonesuppression on insulin-like growthfactor I mRNA in young rats. Journal ofApplied Physiology 76, 2204–2209.

relation to age and adiposity. Journal ofClinical Investigation 67, 1361–1369.

Schlechter, N.L., Russell, S.M., Spencer,E.M. & Nicoll, C.S. (1986) Evidencesuggesting that the direct growth-promoting effect of growth hormone on cartilage in vivo is mediated by localproduction of somatomedin. Proceedingsof the National Academy of Sciences of theUnited States of America 83, 7932–7934.

Schultz, E. (1996) Satellite cell proliferativecompartments in growing skeletalmuscles. Developmental Biology 175, 84–94.

Seale, P. & Rudnicki, M.A. (2000) A newlook at the origin, function, and ‘stem-cell’ status of muscle satellite cells.Developmental Biology 218, 115–124.

Semsarian, C., Wu, M.J., Ju, Y.K et al.(1999) Skeletal muscle hypertrophy is mediated by a Ca2+-dependentcalcineurin signalling pathway. Nature400, 576–581.

Siegfried, J.M., Kasprzyk, P.G., Treston,A.M. et al. (1992) A mitogenic peptideamide encoded within the E peptidedomain of the insulin-like growth factorIB prohormone. Proceedings of theNational Academy of Sciences of the UnitedStates of America 89, 8107–8111.

Singh, M.A., Ding, W., Manfredi, T.J. et al.(1999) Insulin-like growth factor I inskeletal muscle after weight-liftingexercise in frail elders. American Journalof Physiology 277, E135–E143.

Sjogren, K., Liu, J.L., Blad, K. et al. (1999)Liver-derived insulin-like growth factorI (IGF-I) is the principal source of IGF-Iin blood but is not required for postnatalbody growth in mice. Proceedings of theNational Academy of Sciences of the UnitedStates of America 96, 7088–7092.

Vandenburgh, H.H., Karlisch, P., Shansky,J. & Feldstein, R. (1991) Insulin and IGF-Iinduce pronounced hypertrophy ofskeletal myofibers in tissue culture.American Journal of Physiology 260,C475–C484.

Page 210: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

194

Introduction

Fight and flight reaction has been important forhumans throughout history. To be able to reactrapidly and mobilize energy for fast movementsaway from danger, or to be able to exert highamounts of muscle force in confrontations, requiresa system by which a co-ordinated series of eventscan be stimulated by a fast-releasing hormonal sys-tem. The adrenal gland, especially the adrenalmedulla, is very well suited for this. This is becauseepinephrine has potent effects in this regards; the half-life of epinephrine is very short wherebymobilization is quick. Finally, several of the effectsthat epinephrine exerts can be initiated, even in theresting state, when adequate stimulus (mentalstress) can elevate circulating levels of epinephrineto a level that will bring forward, for example,metabolic effects.

Energy turnover increases with onset of exercise,and autonomic/endocrine mechanismsaespeciallythose involving hormones released from the adrenalglandaplay an important role in this regulation.This results in increased mobilization of substrate:i.e. (a) glycogenolysis in skeletal muscle; (b) glucoserelease from the liver to support muscle glycolysis;(c) free fatty acid release from both adipose andmuscle tissue for beta-oxidation in skeletal muscle;and (d) increase protein synthesis. In addition toplaying an important role in substrate mobilizationand combustion, adrenal gland related endocrinemechanisms together with autonomic mechanismscan also: (i) influence blood distribution to variousorgans; (ii) stimulate sweat glands and influence

thermoregulation; (iii) increase contractility of skel-etal muscle; and (iv) cause suppression/stimulationof components involved in the human immune sys-tem during exercise.

Adrenal gland neuroendocrine changeswith exercise

A major component in the autonomic control duringexercise is the adrenergic activity, which can beassessed in humans both by direct measurements ofelectrical activity in superficial sympathetic nervesand by determination of circulating norepinephrineand epinephrine in the blood. Direct recording ofsympathetic activity in humans can, for practicalreasons, be performed to resting muscle only (Searlset al. 1988), whereas direct recording of sympatheticactivity to the adrenal medulla can be determined in animal models. Epinephrine is released from theadrenal medulla in response to sympathetic neuralactivity to the gland during exercise, whereasadrenal cortical hormones (i.e. cortisol) are releaseddue to pituitary hormonal stimulation by adreno-corticotropic hormone (ACTH). Levels of norepine-phrine and epinephrine in arterial blood increasewith exercise intensity, expressed by the percentageof maximal individual performance (% Vo2max),and, as clearance of these hormones only changesmoderately with exercise, changes in plasma levelscan be attributed to changes in secretion and release(Kjær et al. 1985). It has been shown that whole-bodyclearance of epinephrine increases by 15% at low-exercise intensities and decreases around 20% belowbasal levels after more intense exercise (Kjær et al.

Chapter 15

Adrenal Gland: Fight or Flight Implications forExercise and Sports

MICHAEL KJÆR

Page 211: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

adrenal gland and exercise 195

1985). However, as the increase in plasma epine-phrine seen during dynamic exercise in humans isfive to 10-fold, these changes are caused by increasesin secretion from the adrenal medulla rather than by changes in clearance. A major contributor toepinephrine clearance is the hepatosplancnic area aswell as the kidneys.

Epinephrine and cortisol responses are influencedby glucose levels during exercise, and falling glucosemarkedly enhance the responses of these two gluco-counteregulatory hormones during prolonged exer-cise (Galbo et al. 1977). In addition to influencingcarbohydrate and fat metabolism, epinephrine perse has been shown to increase protein metabolism in isolated electrically stimulated muscle. Althoughepinephrine responses can be seen to acute mentalstress, these changes are by far lower than the onesobserved during physical exercise.

Adrenomedullary activity after physical training

Vigorous endurance training will reduce the cate-cholamine response to a given absolute workload,whereas neither sympathetic nerve activity nornorepinephrine levels at maximal workloads differbetween individuals with different training status.This supports the view that physical training doesnot alter the capacity of the sympathetic nervoussystemaand thus most likely neither the sympath-etic activity directed towards the adrenal medullaa

but that responses to submaximal exercise arelinked closely to the relative rather than to the absolute workload. Surprisingly, however, in a 24-hstudy it has been found that, if anything, highlytrained individuals had a higher catecholaminerelease over the day compared with sedentary indi-viduals. Epinephrine response in trained versussedentary individuals has been shown to be enlargedwhen stimulated by a variety of stimuli such ashypoglycemia, caffeine, glucagon, hypoxia andhypercapnia (Kjær et al. 1986, 1988; Kjær & Galbo1988). This indicates that the capacity to secreteepinephrine from the adrenal medulla improveswith training, the development of a so-called ‘sportsadrenal medulla’ reflecting a hypertrophy pheno-menon similar to that seen in a heart adapting to

physical training. This is supported by findings intrained men (sprint) who showed a higher epine-phrine response to short-term exercise comparedwith that of untrained counterparts ( Jacob 2004).Interestingly, no significant difference could beobtained in trained versus untrained women using this short-term sprint test. In rats who underwent 10 weeks of intense swim training, the adrenalmedullary volume and the adrenal content of epine-phrine was larger in trained rats (both male andfemale) compared with controls who were eitherweight matched, sham trained or cold stressed(Stallknecht et al. 1990). Interestingly, the traininginduced increase in adrenal medulla volume wasparalleled by the increase in adrenal gland weight,indicating that the major stimulus was on themedulla rather than the adrenal cortex. Further-more, no increase in total gland weight was seenafter sham training or cold stress, indicating thatthese ‘mental’ types of stress did not result in anymajor gland adaptation. Although these findingsindicate that the improved secretion capacity ofepinephrine is a result of training, this will mostlikely require several years of training. In well-trained athletes who underwent hypoglycemiabefore and 4–5 weeks after an injury that resulted ininactivity, epinephrine responses did not changewith this short-lasting alteration in activity level(Kjær et al. 1992). It could be speculated that an earlyinfluence upon the adrenal medullary volume isneeded, for example during adolescence, and somereports seem to indicate this ( Jacob 2004). However,even if this adaptation requires long-term stimula-tion, it is interesting that endocrine glands appar-ently can adapt to physical training and alter theirsecretion capacity, similar to other tissues like mus-cle and heart.

Motor control and neural reflex influenceon adrenal gland responses

Trying to address the role of motor control foradrenal endocrine responses during exercise canbasically be studied either by enhancing the volun-tary effect to carry out a certain workload and thusexagerating the central effort, or it can be studied bydecreasing the contribution of motor centers by, for

Page 212: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

196 chapter 15

example, performing involuntary exercise and, so tospeak, ‘letting the work being done to you’. Attemptsto increase motor center activity for a given forceoutput has been adressed in humans using partialneuromuscular blockade to weaken the maximalforce by 30–70% (Kjær et al. 1987). It was found thatan exercise-induced increase in levels of circulatingcatecholamines (and pituitary hormones [growthhormone and ACTH]) was augmented compared to control experiments with saline infusion. Thesefindings are supported by experiments in paralyzedcats, where direct stimulation of the subthalamiclocomotor areas in the brain resulted in adrenergichormonal responses similar to the one seen duringvoluntary exercise (Vissing et al. 1989). Together,these experiments support the view that motor center activity can directly stimulate sympatho-adrenergic activity during exercise both directlyand independently of feedback from contractingmuscle. That these central factors coupled to exer-cise intensity are not sufficient to elicit a maximaladrenergic and pituitary hormonal responses can be demonstrated in several ways. When exercisingwith a small muscle group, for example one-leggedknee extension even at maximal intensity, only asmall catecholamine response can be observed(Savard et al. 1989). Furthermore, when maximalwork output was reduced by more than 60% withneuromuscular blockade (tubocurarine), despitesubjects working at the highest possible effort,adrenergic responses were far from maximal.Mechanisms other than central motor control there-fore need to be active during exercise, and one suchmechanism is neural feedback from contracting theskeletal muscle. This can be investigated by usinglumbar epidural anesthesia in doses sufficientlyhigh enough to block impulses in thinner afferentnerves (type III and IV or type C) but still preservingmotor nerves and the ability to perform exercise tothe highest possible degree. Such an approach evid-ently has weaknesses, and negative findings of ablockade does not definitely exclude a role of affer-ent nerves since a perfect distinction between affer-ent and efferent nerves cannot fully be obtained bythis approach. Nevertheless, during static exercise,but not during dynamic exercise, catecholamineresponses were inhibited when afferent responses

were absent. Interestingly, both ACTH and β-endorphin responses during submaximal exercisewere abolished during epidural anesthesia (Kjær et al. 1989). In support of a role of afferent nerves in adrenergic and pituitary responses, hormonallevels in the blood increased in response to directstimulation of these nerve fibers in cats (Vissing et al.1994).

Hepatosplanchnic glucose productionand adrenal gland responses to exercise

During intense exercise the rise in hepatic glucoseproduction parallels a rise in plasma epinephrinelevels. In swimming rats, the removal of the adre-nal medulla reduced the hepatic glycogenolysis.Furthermore, exercise-induced increase in hepatic glucose production was diminished by adreno-demedullation in running rats (Sonne et al. 1985).However, most studies have been unable to demon-strate any direct effect of epinephrine on liver glyco-gen breakdown during exercise. In running dogs,evidence has been provided that epinephrine mayplay a minor role in liver glucose output late duringexercise, probably due to an increased gluconeogenicprecursor level (Moates et al. 1988). Furthermore,adrenalectomized individuals maintain a normalrise in hepatic glucose production during exercise,and only when epinephrine is infused in these pati-ents was hepatic glucose production augmentedduring the early stages of exercise (Howlett et al.1999). In humans the role of liver nerves and epine-phrine have been studied with application of localanesthesia around the sympathetic coeliac ganglioninnervating liver, pancreas and adrenal medulla(Kjær et al. 1993). Pancreatic hormones were stand-ardized by the infusion of somatostatin, glucagonand insulin. During blockade, the exercise-inducedepinephrine response was inhibited by up to 90%,and presumably liver nerves were also blocked, but this did not diminish the glucose productionresponse to exercise. This indicates that sympatho-adrenergic activity is not responsible for the exercise-induced rise in splanchnic glucose output. In furthersupport of this hypothesis, the exercise-inducedincrease in liver glucose production was identical inliver-transplanted patients compared to healthy

Page 213: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

adrenal gland and exercise 197

control subjects, as well as kidney-transplantedpatients who received a similar hormonal andimmunosuppressive drug treatment as the liver-transplanted patients (Kjær et al. 1996a). Finally, inexperiments in exercising dogs that underwent aselective blockade of hepatic α- and β-receptors, itwas demonstrated that circulating norepinephrineand epinephrine do not participate in the stimula-tion of glucose production during intense exercise(Coker et al. 1997). Taken together, sympathetic livernerves or circulating norepinephrine play no role inglucose mobilization from the liver during exercise,and circulating epinephrine only plays a minor roleduring intense exercise and late, prolonged exercise.Cortisol has been shown only to play a minor role inhepatic glucose production during exercise, andonly seems to be of importance if inadequate secre-tion of other hormones is present.

Epinephrine effect on musclecarbohydrate metabolism

Epinephrine has been demonstrated to stimulateglycogen breakdown in skeletal muscle during contraction both in exercising animals and humanswhen supra-physiological doses were used (Richter1996). Later studies using more physiological epine-phrine doses have not been able to demonstrate any increased glycogen breakdown despite higheractivation of phosphorylase compared with controlstudies). In line with this, in adrenalectomized indi-viduals no impairment in glycogen degradation wasfound during exercise, neither was muscle glyco-genolysis increased by substituting epinephrineduring exercise (Kjær et al. 2000). It was furthermoreshown that activation of glycogen phosphorylaseand hormone-sensitive lipase is only present if theseindividuals receive infusion with epinephrine tomimic changes in epineprine occuring in healthyindividuals during exercise. This indicates the im-portance of epinephrine in the activation of glyco-genolytic and lipolytic pathways. It also indicatesthat such activation occurs in parallel for intramuscu-lar triglyceride and glycogen by adrenergic activity,and that the choice of substrate for energy produc-tion takes place at another level in the muscle (Kjæret al. 2000).

Spinal-cord-injured individuals are characterizedby a lack of voluntary control over their lower limbs,and are absent of neural feedback from muscles to higher brain centers. Development of equipmenthas allows for functionally electrical-stimulatedexercise on an ergometer, causing oxygen uptakerates to rise to around 1.0–1.5 L·min–1. This allows forthe study of carbohydrate and fat metabolism andmetabolic changes during exercise. Using involunt-ary exercise in spinal-cord-injured individuals as anintervention showed that, in the absence of motorcontrol and neural feedback from muscle, mobiliza-tion of glucose from the liver was impaired, resultingin a gradual drop in plasma glucose during exercise(Kjær et al. 1996b). In line with this, healthy indivi-duals who were paralyzed by an epidural blockadealso had impaired glucose mobilization response(Kjær et al. 1998). Furthermore, in spinal-cord-injured patients during voluntary arm cranking,euglycemia was maintained during exercise. Thesefindings indicate that neural mechanisms are crucialfor the matching of glucose mobilization to periph-eral glucose uptake during exercise, and that blood-borne mechanisms are not sufficient to accomplishthis. During electrical exercise by spinal-cord-injuredindividuals, the primary energy source is glyco-genolysis, and higher levels of lactate have beenfound both in muscle and blood. Furthermore, theglucose uptake is several-fold higher in spinal-cord-injured patients compared with healthy controlsworking at the same oxygen uptake rate.

Sympathoadrenergic activity and fat metabolism

Intravenous infusion of epinephrine in restinghumans caused an increase in lipolytic activity as determined by microdialysis of subcutaneous adipose tissue, an effect that was desensitized byrepeated epinephrine infusions (Stallknecht 2003).Spinal-cord-injured patients were investigated dur-ing arm cranking, and lipolysis was determined bymicrodialysis in subcutaneous adipose tissue bothabove and below the cutaneous border separat-ing the sympathetic innervated region (proximal–clavicular region) from the desympathectomizedareas (distal–umbilical region) (Stallknecht et al.

Page 214: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

198 chapter 15

2001). In both regions lipolysis increased with exer-cise, evidence that direct sympathetic innervation isnot of major importance for the lipolysis duringmuscular work. In contrast, circulating epinephrinemay be a likely candidate for lipolysis activation.Training causes a decreased adipose tissue mass andadipocyte size, but the sympathoadrenergic systemdoes not seem to be crucial for this adaptation.

Not only adipose tissue but also intramuscular fatcan be stimulated by epinephrine, and both lipopro-tein lipase (LPL) and hormone sensitive lipase(HSL) play important roles in this regulation. HSLmight be under control by both contractions andepinephrine (Donsmark 2002), and it has recentlybeen shown that activation of HSL and glycogenphosphorylase occurs in parallel in adrenalectom-ized individuals who receive an infusion withepinephrine during exercise (Kjær et al. 2000). Thiscould indicate that the mobilization of intramuscu-lar triglyceride and glycogen occurs when simul-taneously stimulated by adrenergic activity, and

that the choice of substrate for energy productiontakes place at another level.

Summary

The adrenal gland plays an important role in releas-ing hormones that are crucial for carbohydrate, fat and protein metabolism, as well as for regu-lating organ blood distribution, thermoregulation,immunmodulation and skeletal muscle contractilityduring and after exercise. Epinephrine secretionincreases with the relative work intensity, andadrenal medulla hypertrophy can occur in responseto prolonged intense training (‘sports adrenalmedulla’). Both direct motor center activity andafferent neural feedback signaling from contractingmuscle play a role in the regulation of hormonalrelease from the adrenal gland during exercise.Adrenal gland hormones (epinephrine and cortisol)participate in the redundant regulation of hepaticglucose release during physical activity.

References

Coker, R.H., Krishna, M.G., Brooks Lacy,D., Bracy, D.P. & Wasserman, D.H.(1997) Role of hepatic α- and β-adrenergic receptor stimulation onhepatic glucose production duringheavy exercise. American Journal ofPhysiology 273, E831–E838.

Donsmark, M., Langfort, J., Ploug, T. et al. (2002) Hormone-sensitive lipase(HSL) expression and regulation byepinephrine and exercise in skeletalmuscle. European Journal of Sports Science2, 1–10.

Galbo, H., Christensen, N.J. & Holst, J.J.(1977) Catecholamines and pancreatichormones during autonomic blockade in exercising man. Acta PhysiologicaScandinavica 101, 428–437.

Howlett, K., Lorentsen, J., Bergeron, R. et al. (1999) Effect of adrenaline onglucose kinetics during exercise inadrenalectomized subjects. Journal ofPhysiology 519, 911–919.

Jacob, C., Zouhal, H., Prioux, J. et al. (2004)Effect of the intensity of training on catecholamine responses tosupraphysiological exercise inendurance-trained men. EuropeanJournal of Applied Physiology 91, 35–40.

Kjær, M. & Galbo, H. (1988) The effect ofphysical training on the capacity tosecrete epinephrine. Journal of AppliedPhysiology 64, 11–16.

Kjær, M., Christensen, N.J., Sonne, B.,Richter, E.A. & Galbo, H. (1985) Effect of exercise on epinephrine turnover intrained and untrained male subjects.Journal of Applied Physiology 59,1061–1067.

Kjær, M., Farrell, P.A., Christensen, N.J. &Galbo, H. (1986) Increased epinephrineresponse and inaccurate glucoregulationin exercising athletes. Journal of AppliedPhysiology 61, 1693–1700.

Kjær, M., Secher, N.H., Bach, F.W. &Galbo, H. (1987) Role of motor centeractivity for hormonal changes andsubstrate mobilization in exercisingman. American Journal of Physiology 253,R687–R695.

Kjær, M., Bangsbo, J., Lortie, G. & Galbo, H. (1988) Hormonal response toexercise in man: influence of hypoxiaand physical training. American Journal of Physiology 254(23), R197–R203.

Kjær, M., Secher, N.H., Bach, F.W., Sheikh,S. & Galbo, H. (1989) Hormonal andmetabolic responses to exercise in

humans: effect of sensory nervousblockade. American Journal of Physiology257, E95–E101.

Kjær, M., Mikines, K.J., Linstow, M., Galbo, H. & Nicolaisen, T. (1992) Effectof 5 weeks detraining on epinephrineresponse to insulin inducedhypogycemia in athletes. Journal ofApplied Physiology 72(3), 1201–1204.

Kjær, M., Engfred, K., Fernandes, A.,Secher, N.H. & Galbo, H. (1993)Regulation of hepatic glucoseproduction during exercise in man: role of sympathoadrenergic activity.American Journal of Physiology 265,E275–E283.

Kjær, M., Keiding, S., Engfred, K. et al.(1996a) Glucose homeostasis duringexercise in humans with a liver orkidney transplant. American Journal ofPhysiology 268, E636–E644.

Kjær, M., Pollack, S.F., Mohr, T. et al.(1996b) Regulation of glucose turnoverand hormonal responses duringexercise: electrical induced cycling intetraplegic humans. American Journal ofPhysiology 271, R191–R199.

Kjær, M., Secher, N.H., Bangsbo, J. et al.(1998) Hormonal and metabolic

Page 215: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

adrenal gland and exercise 199

responses to electrically induced cyclingduring epidural anesthesia in humans.Journal of Applied Physiology 80,2156–2162.

Kjær, M., Howlett, J., Langfort, T. et al.(2000) Adrenaline and glycogenolysis inskeletal muscle during exercise: a studyin adrenalectomised humans. Journal ofPhysiology 528, 371–378.

Moates, J.M., Lacy, D.B., Cherrington, A.D.,Goldstein, R.E. & Wasserman, D.H.(1988) The metabolic role of the exercise-induced increment in epinephrine.American Journal of Physiology 255,E428–436.

Richter, E.A. (1996) Glucose utilization. In: Handbook of Physiology (Rowell, L.B. & Shepherd, J.T., eds.). Bermedica,Columbia, MD: 912–951.

Savard, G., Richter, E.A., Strange, S. et al.(1989) Noradrenaline spillover during

despite enlarged adrenal medulla intrained rats. American Journal ofPhysiology 259, R998–R1003.

Stallknecht, B. Enevoldsen, L., Lorentsen, J. et al. (2001) Role of thesymphathoadrenergic system in adiposetissue metabolism during exercise inhumans. Journal of Physiology 536,283–294.

Vissing, J., Iwamoto, G.A., Rybicki, K.,Galbo, H. & Mitchell, J.H. (1989)Mobilization of glucoregulatoryhormones and glucose by hypothalamiclocomotor centers. American Journal ofPhysiology 257, E722–E728.

Vissing, J., Iwamoto, G.A., Fuchs, I.E.,Galbo, H. & Mitchell, J.H. (1994) Reflexcontrol of glucoregulatory exerciseresponses by group III and IV muscleafferents. American Journal of Physiology266, R824–R830.

exercise in humans: role of muscle mass.American Journal of Physiology 257,H1812–1818.

Searls, D.R., Victor, R.G. & Mark, A.L.(1988) Plasma norepinephrine andmuscle sympathetic discharge duringrhythmic exercise in humans. Journal ofApplied Physiology 65, 940–944.

Sonne, B., Mikines, K.J., Richter, E.A.,Christensen, N.J. & Galbo, H. (1985) Roleof liver nerves and adrenal medulla inglucose turnover of running rats. Journalof Applied Physiology 59, 1640–1646.

Stallknecht, B. (2003) Influence of PhysicalTraining on Adipose Tissue Metabolism—with Special Focus on Effects of Insulin andEpinephrine. PhD thesis. University ofCopenhagen, Denmark.

Stallknecht, B., Bülow, J., Frandsen, E. & Galbo, H. (1990) Diminishedepinephrine response to hypoglycemia

Page 216: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

200

The adrenal gland

The two endocrine adrenal glands are relativelysmall glands that are situated superior to the kidney.These glands are essential for life; removal of them would result in death. Diseases of the adrenalglands occur but are generally treated with syn-thetic derivatives of the hormones these glands pro-duce. The main function of the adrenal glands is to respond to stressed of the body by producing hormones that regulate blood glucose, increaseheart rate and ventilation, and maintain the fluidhomeostasis in the body. The adrenal gland has thehighest rate of blood flow in the body per gram oftissue, and is comprised of two main types of tissue;the cortex and medulla (Kaplan 1988). The adrenalmedulla is the innermost part of the adrenal glandand comprises only about 10% of the total gland tis-sue. The adrenal medulla is a sympathetic ganglionin which postganglionic fibers have ended in secret-ory vesicles. Upon nervous system stimulation, viapreganglionic nerves reaching the adrenal medullavia the splanchnic nerve, hormones and neurohor-mones (i.e. epinephrine and peptide F) are secretedfrom chromaffin cells. Adrenal medullary hormonesare not essential for life but are produced andsecreted during stressful-like events. The adrenalcortex, on the other hand, is essential for life. Thisouter cortex layer secretes glucocorticoids, involvedin carbohydrate and protein metabolism (i.e. cor-tisol); mineralcorticoids, involved in maintenance of extracellular fluid volume (i.e. aldosterone); andminor-effecting sex hormones involved in repro-duction. The adrenal cortex is stimulated primarily

by the anterior pituitary release of adrenocortico-tropic hormone.

Adrenal medullary chromaffin system

The adrenal medulla contains cells known as chro-maffin cells which constitute the majority of spacewithin the medulla. The chromaffin system servesas the method of storage and secretion for cate-cholamines and proenkephalin peptide fragments,including peptide F, E, and B (Lewis et al. 1979;Viveros et al. 1979). They resemble storage andsecretory granules of other endocrine or exocrineglands found within the body. These chromaffingranules are highly specialized, electron-dense andosmiophilic organelles smaller than mitochondria.Communication between the sympathetic chromaf-fin system and the body is rapid, where the neuralresponse is instantaneous and the hormonal res-ponse occurs within minutes or hours. Stimulationof chromaffin cells by sympathetic preganglionicneurons of the splanchnic nerve (Coupland 1965,1972) occurs in response to the neurotransmitteracetylcholine secreted from preganglionic neuronsarising from the thoracolumbar region of the spinalcord (Fig. 16.1).

Besides enkephalins and catecholamines, someother constituents of the chromaffin granules existincluding chromogranin A, nucleotides and ascor-bic acid. Extra-adrenal chromaffin cells are locatedadjacent to the aorta as paraganglia, in carotid bodies, in viscera, and in sympathetic ganglia. Todate, there is only indirect evidence for selective dis-charge of granular contents upon neural stimulation

Chapter 16

The Adrenal Medulla: Proenkephalins andExercise Stress

JILL A. BUSH AND N. TRAVIS TRIPLETT

Page 217: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 201

of the adrenal medulla. However, exocytosis of the chromaffin granule is far from simple, and thisprocess could provide a potential mechanism forthe differential release pattern with exercise stress.Unfortunately, there is no data available to supportexocytosis and hence the differential release patternof adrenal medullary neurohormone (i.e. peptide Fand epinephrine) upon stimulation. Stimulation ofnicotinic and muscarinic receptors is involved inacetylcholine-related exocytosis of the chromaffincells. Several endogenous substances (e.g. substanceP, neurotensin, vasoactive intestinal peptide) withinthe adrenal medulla may competitively bind tonicotinic receptors, thus inhibiting the release ofepinephrine (Livett, B.G. 1984). High concentrationsof nicotine demonstrated a proportional release ofenkephalin-containing peptides and total catechola-mine concentration (Wilson et al. 1982; Livett, B.G.1984). If exocytosis of chromaffin cells containingproenkephalin fragment peptide F and epinephrinecan be altered by such endogenously produced sub-stances, then this would provide a plausible explana-tion for a differential release pattern of peptide Fand epinephrine.

Chromaffin granules contain membrane-boundproteins and glycoproteins. Such soluble factorsinclude a proton pump adenosine triphosphatase(ATPase), catecholamine and nucleotide carrier proteins, cytochrome b561, actin, glycoproteins, anddopamine β-hydroxylase pump ATPase is a neces-

sary constituent of the chromaffin granule (Fig. 16.2).Via hydrolysis of ATP on the exterior membranesurface, protons (H+) are injected into the granule,providing a more acidic, positively charged envir-onment versus the cytosol. An electrochemical H+

gradient is created in the interior of the granule. Dueto the biosynthetic pathways of epinephrine anddopamine, the energy supplied by the electrochem-ical proton gradient is necessary for the uptake of these two substances into the cell. The enzymenecessary to breakdown norepinephrine into epine-phrine is located in the cytosol of the cell. Thus nore-pinephrine must be exported from the cell, exposedto the enzyme, and epinephrine recaptured for storage inside the granule. Other factors may also beinvolved in the exocytotic process of the granules. A Ca2+ influx is a primary elicitor of exocytosis.Stimulation via acetylcholine or high extracellularK+ increases the intracellular Ca2+ concentration ofthe chromaffin cells. As the granules move towardthe plasma membrane, it may encounter the cyto-skeletal structure actin. The increase in Ca2+ maydecrease the viscosity of this interaction permittingeasier granular movement. Calmodulin selectivelyphosphorylates membrane proteins, allowing forthe fusion of plasma and granular membranes.Synexin, in the presence of Ca2+, can polymerizeinto large aggregates which bind to phospholipidswithin the membrane. Also, recycling of the granu-lar membrane to and from the plasma membrane is

Spinalcord

Prevertebralchain ganglia

Splanchnicnerve

Adrenalgland

Kidney

Medulla

Chromaffin cells

Epinephrine PFEpiPF

Ach

20%

Cortex

80%

Fig. 16.1 Neural innervation andchromaffin granules of the adrenalgland. Ach, acetylcholine; Epi,epinephrine; PF, peptide F.

Page 218: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

202 chapter 16

continuously occurring. All of these aforementionedprocesses are important in the role of exocytosis of the chromaffin granules containing epinephrineand peptide F in the differential release of eitherneurohormone.

The chromaffin cells also secrete enkephalins, a group of substances that possess morphine-likeanalgesic activity and are also produced by the brain(Undenfriend & Kilpatrick 1984). The enkephalinsand some larger peptide fragments which containenkephalin sequences, known as the enkephalin-containing peptides (ECPs), are derived from thelarge molecular weight preproenkephalin. TheseECPs include peptides in the 3–5 kDa range, whichwould include peptides E, F and B (Undenfriend &Kilpatrick 1984; Hiddinga et al. 1990). Studies withthese peptides have found parts of their amino acidsequences to be highly conserved among differentspecies (Lewis & Stern 1983) and are found in theblood in physiologically relevant concentrationswith long half-lives (≥ 15 min), indicating their bio-logical significance (Katzenstein et al. 1987). How-ever, many of the functions of these substances areas yet unknown.

Proenkephalin peptide F

It is important to understand some of the funda-

mental aspects of the proenkephalin polypeptideprecursor and the processing of this peptide precur-sor to peptide F in order to appreciate the potentialphysiological roles of peptide F. The preproen-kephalin polypeptide (30 kDa) is the precursor ofthe enkephalins ([Met]- and [Leu]-enkephalin) andECPs and is found in various regions of the body:brain, adrenal medulla and activated T-lymphocytes,and hemoglobin molecules (Hughes et al. 1975;Kimura et al. 1980; Brantl et al. 1986; Zurawski et al.1986; Martin et al. 1987; Ivanov et al. 1997; Zhao et al.1997).

Peptide F, a 3.8 kDa ECP, is post-translationallyprocessed from preproenkephalin (Fig. 16.3) in aseries of cleavages by trypsin (amino end) and car-boxypeptidase B (carboxyl end) which cleave at theenkephalin sequences (Undenfriend & Kilpatrick1984). Peptide F, the more common peptide productof proenkephalin precursor measured in circulation(Kilpatrick et al. 1980, 1981; Wasserman et al. 1986),consists of 34 amino acids (amino acid sequence 107–140) and contains two [Met]-enkephalin sequences(Lewis 1982; Lewis & Stern 1983), expressing struc-tural similarities to classical opiates (Hansen &Morgan 1984). Opioid peptides typically play anessential role in the brain as neurotransmitterswhere [Met]- and [Leu]-enkephalin are the mostprominent end-products.

Ca2+

Ca2+ channelCa2+

Achstimulation

+CaM

inactive

Catecholaminesynthesis and

secretion

Phosphorylate several proteins Chromogranin A Tyrosine hydroxylase Chromaffin granule membrane Others

Achrec

Ca2+/CaMactive

CaMkinase II

Cell membrane

Fig. 16.2 Signal transductionpathway in acetylcholine-stimulatedadrenal medullary cells. Ach,acetylcholine; CaM, calmodulin.(Modified from Yanagihara et al.1996.)

Page 219: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 203

Interestingly, a breakthrough in the discovery ofthe preproenkephalin precursor molecule becameapparent when Kimura et al. (1980) reported infor-mation pertaining to possible precursors for theenkephalins, [Met]- and [Leu]-enkephalin, secretedfrom the adrenal medulla. With the discovery of few fragments in the brain, the determination of the structure of the precursor became difficult. ECPs derived from the biosynthetic processing ofproenkephalin within the central nervous systemcannot fully account for the total amount of thesepeptides found in the circulatory pool (Lewis &Stern 1983). However, a higher concentration of[Met]- and [Leu]-enkephalin is derived throughcentral nervous system processing (Lewis 1982;Lewis & Stern 1983). Discovery of more proenke-phalin fragments in the adrenal medulla gave rise to better biochemical determination of sequences of the preproenkephalin precursor and to potentialphysiological stress responses. It was also thoughtat the time that the more labile enkephalins weretransported via the higher molecular weight frag-ments (e.g. to peptide F) since ECPs have a greater

circulating period (i.e. 15–60 min) than the smaller[Met]- and [Leu]-enkephalins (i.e. 1–2 min) (Kilpatricket al. 1980, 1981; Kimura et al. 1980; Boarder &McArdle 1986).

Transportation of 3–8 kDa peptides through thecirculation could be enhanced by the non-enkephalinregions within the peptide structure (Boarder &McArdle 1986), thus adding to the prolonged plasmahalf-life of these peptide fragments. The roles of the [Met]- and [Leu]-enkephalin in the peripherybecame speculative at best due to a high potentialfor protease degradation, within 1–2 min (Kraemeret al. 1987). Thus, the larger structure of peptide Fwith a longer prolonged circulating half-life (15–60 min) provided the potential for physiologicalroles in the peripheral circulation (e.g. communica-tion between different biocompartments).

Concomittant release of peptide F andepinephrine from adrenal medulla

Peptide F is co-stored with epinephrine within chro-maffin cells of the adrenal medulla (Viveros et al.

Met-enkephalin

Leu-enkephalin

Met-enk–Arg–Phe

Met-enk–Arg–Gly–Leu

Preproenkephalin: 29.8 kDa

18.2 kDa 4.9 kDa

Peptide I:

Peptide E:

Peptide F:

Peptide B:

2.6 kDa

12.6 kDa 5.3 kDa 3.2 kDa

8.6 kDa 3.8 kDa

Signalpeptide

Fig. 16.3 Post-translationalprocessing of enkephalin-containingpeptides from preproenkephalin.(Modified from Lewis & Stern 1983,Undenfriend & Kilpatrick 1984 andKatzenstein et al. 1987.)

Page 220: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

204 chapter 16

1979; Lewis & Stern 1983). However, the biochem-ical and molar-equivalent relationship of epineph-rine and peptide F within these chromaffin cells isless understood. The relationship of the co-storage ofpeptide F and epinephrine may provide a potentialmechanism for differential release of these neuro-hormones from the adrenal gland in response tostress. It is known that trypsin and carboxypepti-dase β digestion of preproenkephalin within theadrenal medulla occurs within 2 h of proenkephalinprecursor synthesis and that the peptide F fragmentis found in the chromaffin granules within 2 h of theprecursor synthesis (Wilson 1991). Tetrabenazine,which has been shown to inhibit catecholamineuptake into the storage vesicles, did not inhibituptake of proenkephalin fragments into vesicles. Infact, it enhanced the peptide’s assimilation (Wilson1991). This research would suggest that, not only ispeptide F a final processing product assimilatedinto the chromaffin vesicles, but also that termina-tion of the process may depend upon the co-storageof epinephrine (Wilson 1991).

Studies have shown that epinephrine and enke-phalin-containing fragments are co-released fromcultured adrenal chromaffin granules (Schultzberget al. 1978; Livett, A.R. et al. 1981). This co-secretion,under similar physiological stimulation, also under-scores the importance of a biological role for peptideF during stress. It is also possible that peptide F andepinephrine can have a complementary action onvarious biological target tissues. For example, bothepinephrine (McCarthy & Dale 1988) and peptide F (Hiddinga et al. 1994) have modulating effects on the immune system. This may be similar to amodel used by other neurohormones and peptideswhich are co-stored and co-secreted and possessdissimilar/similar biological activities (e.g. insulinand serotonin within pancreatic β-cells in humans[Richter et al. 1986] and neuropeptide Y and nor-epinephrine within bovine sympathetic nerves[Bastiaensen et al. 1988; De Potter et al. 1988]).

Epinephrine is the primary stress hormone in thebody. Its biosynthesis and release may influencepeptide F release and appearance in the blood(Kraemer et al. 1985b, 1991). While it is important tounderstand the processing and storage of peptide F, it is important to understand the processing of

epinephrine as well, as a close relationship existsbetween these neurohormones. The enzymatic pro-cessing of newly synthesized preproenkephalinmay be affected by epinephrine and thus may affectthe co-storage of peptide F (Wilson 1991). Wilsonand colleagues (Wilson et al. 1982; Wilson 1991)found that pharmacological agents and neuro-transmitters activated proenkephalin synthesis inthe chromaffin cells. They found that inhibitors ofvesicular catecholamine uptake (e.g. tetrabenazineand resperine) enhanced the processing of proenke-phalin and thus the content of ECPs in the chro-maffin cells. This data suggested that epinephrine or possibly other constituents of the chromaffin cells(e.g. adenosine triphosphate [ATP]) may inhibitproenkephalin processing. Thus, this mechanism ofaction can alter the molar concentration of peptide Fand epinephrine within different chromaffin cells.

It is important to understand the basic processingof epinephrine. Briefly, amines containing a 3,4-dihyroxyphenyl nucleus are referred to as catecho-lamines and are derived from the amino acidtyrosine. Tyrosine can also be derived from the conversion of phenylalanine in the liver via pheny-lalanine hydroxylase. Tyrosine and phenylalaninecan be found in the diet of foods high in protein.Epinephrine is mainly derived from the adrenalmedulla; however, the necessary enzymes involvedin biosynthesis are also present within neurons ofthe central nervous system in minute amounts.Norepinephrine is primarily found within the cent-ral nervous system as a neurotransmitter for sym-pathetic neurons, either inhibitory or excitatory.Approximately 80% of total stimulated adrenalmedullary catecholamine secretion is epinephrine,while the remaining 20% is norepinephrine.

The initial step in epinephrine synthesis involvesconversion of tyrosine to 3,4-dihydroxyphenylala-nine (dopa) via the enzyme tyrosine hydroxylase,which is phosphorylated and activated when stimu-lated by acetylcholine (Fig. 16.4). Dopa, in the pres-ence of an aromatic l-amino acid decarboxylase, isdecarboxylated to form dopamine. The two afore-mentioned enzymes are located in the cytosol of the medulla where such enzymatic reactions occur.Dopamine enters the granule and is hydroxylatedby dopamine β-hydroxylase from the granular mem-

Page 221: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 205

brane to form norepinephrine. Norepinephrinereturns to the cytosol for methylation into epineph-rine via phenylethanolamine N-methyl transferase(PNMT), using S-adenosyl-l-methionine as a donor.Epinephrine is reincorporated into the granule forstorage and secretory preparation. This process isdependent on the energy from the proton pumpATPase. Catecholamines within the granule arebound to ATP and chromogranin A, preventingegress of the stored hormones.

In the periphery (i.e. adrenal gland), epinephrine,norepinephrine and dopamine serve as neurohor-mones, while in the central nervous system, theycan serve as neurotransmitters. The plasma half-lifeof epinephrine is about 1–2 min. The plasma concen-tration of epinephrine at rest is ~ 0.05 ng⋅mL–1 andcan elevate from 0.27 ng⋅mL–1 to 4.1 ng⋅mL–1 duringexercise. Epinephrine affects the state of arousal, the‘fight or flight’ response, and the contractility ofheart and skeletal muscle. Epinephrine elevatesheart rate, increases blood flow to skeletal muscle,increases the metabolic rate, and increases substrateutilization during exercise via release of glucose andfree fatty acids into the blood. Epinephrine binds to receptors on the cell surface of the plasma mem-brane of target cells, interacting with both α- (α1, α2)

and β- (β1, β2, β3) adrenergic receptors. Epinephrinehas a high affinity for β2-receptors which are locatedextrajunctionally on non-innervated target cells.Such receptors mediate lactate production andvasodilation in skeletal muscle. The role of epine-phrine release during acute exercise stress mayobviate the role played by peptide F, as the patternof response between the two neurohormones some-times differs (Kraemer et al. 1985b, 1991). This is evident when data are expressed relative to themolar ratio of peptide F to epinephrine. A decreasein the molar ratio (usually observed during exer-cise) would indicate the predominance of epine-phrine secretion into the circulation, whereas asincrease in the molar ratio (usually observed duringrecovery) would indicate the predominance of pep-tide F secretion. However, it cannot be discountedthat different chromaffin cells contain varying con-centrations of each neurohormone (Wilson et al.1982; Livett, B.G. 1984), or that chromaffin cells areselectively released upon stimulation. Epinephrinehas been shown to play a greater role during exer-cise (Kjær et al. 1985; Brooks et al. 1988; Kjær & Galbo 1988), while peptide F may play a greater role during recovery from exercise (Kraemer et al. 1985b,1991). The difference in release patterns may explainimportant biological functions. The possibility ex-ists that, even though epinephrine and peptide F areco-stored and co-secreted by similar stimuli, theymay have different physiological roles within thesame biocompartment.

Physiological role of peptide F

Although peptide F contains two [Met]-enkephalinsequences, it responds only weakly to classical opiate action tests (Lewis & Stern 1983). Therefore,the [Met]-enkephalin sequence contained in peptide F is not necessarily indicative of enkephalin-likefunctions. The non-enkephalin segments of the pep-tide F molecule have an important role in their func-tion, as they may be the primary binding sequencesfor the molecule. Figure 16.5 outlines potentialphysiological roles for peptide F that will be dis-cussed in the following section.

To date, one of the most important evidences supporting the study of the response pattern of

Diet

GI tractTyrosine

Dopa

Dopamine

Norepinephrine

Epinephrine

Tyrosine hydroxylase

Phenylalaninehydroxylase

Liverconversion

Vascularcompartment

PhenylalanineTyrosine

Aromatic L-amino aciddecarboxylase(cytoplasm)

Dopamine β-hydroxylase(chromaffin granules)

PhenylenthanolamineN-methyl transferase(cytoplasm)

Fig. 16.4 Biosynthetic pathway of adrenal medulla-derived epinephrine. (Modified from Genuth 1988.)

Page 222: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

206 chapter 16

peptide F is the interaction between peptide F andthe immune system (Hiddinga et al. 1994; Triplett-McBride et al. 1998). In addition, it is possible thatthese interactions can take place in the blood bio-compartments. Peptide F has been shown to have a relationship with T- and B-cells in vitro (Hiddingaet al. 1994) and to be related to B-cell activation andindividual fitness level (Triplett-McBride et al. 1998).

The longer half-life of peptide F prolongs its bio-logical viability to interact with the different bloodbiocompartments and the immune system. How-

ever, the immune system may not be the only targetsite for this proenkephalin peptide F fragment.Other target tissues and sites may exist as this is anevolving form of study.

Hiddinga et al. (1994) reported a possible role forpeptide F as a regulatory neurohormone within theimmune system. Unlike the suppressive effects of[Met]-enkephalin on the immune system (Johnsonet al. 1982; Marotti et al. 1993), peptide F seems toenhance immune function (Hiddinga et al. 1994). Invitro research using purified peptide F at physiolo-

Splanchnic nervestimulation

Secretion of epinephrineand peptide F into bloodfollowing exocytosis ofchromaffin cells

Epinephrine

α β receptors

Metabolic effectson tissues in body

HR

Peptide F

Adrenal medulla

Chromaffin cellscontaining variousmolar concentrationsof epinephrineand peptide F

Circulate 15 minin plasma

Cleaved into peptide F?

Secrete proenkephalin

Immunecells

RBC

RBC

Potential interactionwith immune system

Secrete Hbopiate-likefragments

Autocrinefeedback topotentialopiate-likereceptors

Potential interactionwith erythrocyte

opiate-like receptors

Enhance immune function

Epinephrine only

ContractilityGlycogenolysisGluc utilization

Peptide F only

Combination

Fig. 16.5 Potential interaction of peptide F within the three biocompartments of blood. HR, heart rate; RBC, red blood cell.(From Bush et al. under review.)

Page 223: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 207

gical concentrations demonstrated that, following15 min of peptide F incubation with T-cells, therewas an increase in antibody-forming B-cells in cul-ture, significantly enhancing the antigen-specificantibody-forming cell response of lymphocytes toan antigenic challenge of trinitrophenyl-Ficoll (TNP-Ficoll) (51 ng⋅mL–1). Studies in athymic nude mice(e.g. mice expressing delayed and defective T-cell development) demonstrated that peptide F was directly involved in an alteration of the cell-mediated immune response (i.e. T-cell activation)rather than directly involved in the humoral-medi-ated immune response (i.e. antibody productionfrom B-cells). This data has shown that lymphocytescould be a possible target site for peptide F, and thatsubsequent interaction causes an enhancement inimmune function.

Methods of identifying the peptide F receptor

Competitive binding with naloxone. Hiddinga et al.(1994) also attempted to indirectly identify a pos-sible receptor mechanism for peptide F throughnaloxone competition. Naloxone functions as anantagonist for opiate receptor binding (e.g. [Met]-enkephalin) (Simonds 1988). Murine splenocyteswere either treated with peptide F or [Met]-enkephalin to determine which molecule had thegreatest effect on the antibody-forming cell responseof B-cells. Naloxone was added at final concentra-tions of 0.1, 1.0 and 10.0 µmol (i.e. 10–1000 timesgreater than the concentration of either plasma peptide F or plasma [Met]-enkephalin). At a 10 nmolconcentration, purified peptide F and [Met]-enkephalin, respectively, yielded enhancement andsuppression on the antibody-forming response of B-cells. To activate the lymphocytes, antigens (1%sheep red blood cells [SRBC] or TNP-Ficoll) wereadded to the culture at both an optimal (51 ng⋅mL–1)and suboptimal (5 ng⋅mL–1) concentration follow-ing 15 min of incubation with naloxone (10 µmol)and/or peptide F (10 nmol). Naloxone was unableto block the antibody-forming response of B-cellselicited by peptide F. This indicated that peptide Fmay interact with a yet-to-be-determined receptorother than an opioid receptor (i.e. a peptide receptor)on lymphocytes. [Met]-enkephalin-induced suppres-

sion of the antibody-forming response of B-cells, and this action was blocked by the addition of naloxone. This suggested that this low molecularweight enkephalin was bound to opiate receptorsfound on lymphocytes (Sibinga & Goldstein 1988).To test if concentration of either molecule was animportant factor in modulating immune function,peptide F and [Met]-enkephalin were added simul-taneously to cultures in an equimolar concentration(10 nmol) in the presence of naloxone. [Met]-enkephalin again induced suppression of the anti-body-forming response of B-cells, and this actionwas blocked by naloxone. Peptide F, on the otherhand, induced an increase in antibody production,and this increase was not blocked by naloxone. Thisagain suggested the possibility of different mech-anisms of receptor binding on the same immunecells. It may be that naloxone inhibited the immuno-suppressive effects of [Met]-enkephalin, therebypotentiating the immunoenhancing effects of pep-tide F. These data also implied that even though thepeptide F molecule contained [Met]-enkephalinsequences, peptide F may not have interacted withopioid receptors found on lymphocytes. It may bethat the non-enkephalin (peptide) sequences of pep-tide F play a greater role in receptor binding (Roth et al. 1989). This line of research has proven to bevery beneficial in determining and locating a pos-sible receptor and/or target site for peptide F, whichto date are not fully defined. Such data and the rela-tionship of immune cells and peptide F supportedthe need for research since peptide F was found inquantifiable concentrations in the white blood cellbiocompartment of the blood.

Immunochemical approach via flow cytometry. Sincethese studies in the 1980s, Bush et al. (under review)have performed other methodological techniques to determine the receptor for peptide F on immunecells. It was reasoned that an immune cell respons-ive to peptide F would bear peptide F receptors. An immunochemical approach was used to concur-rently identify cells displaying a peptide F receptorand to identify the leukocyte subtype. Monoclonalantibodies to be used for identification of subclassesof human leukocytes were commercially available.No antibodies were available to identify peptide F

Page 224: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

208 chapter 16

receptors. In fact, peptide F receptors have not beencharacterized. Therefore, an indirect approach andflow cytometry was employed (Fig. 16.6).

An antibody to the ligand (i.e. peptide F) wasused to determine a receptor identified by ligandbinding. A series of experiments including numer-ous controls to optimize the system and to verifyspecificity were performed. Such experiments in-cluded variation of the dose of peptide F and anti-peptide F antibody; utilization of several blockingbuffers to eliminate background staining; and test-ing in the absence of exogenous peptide F, primaryantibody, and secondary antibody. Normal rabbitserum (several different animals) was used as a neg-ative control. Dual labeling techniques were used totry to localize the labeling to a subclass of leukocytes.

Using the indirect approach (Fig. 16.6), it wasfound that between 20–30% of the white blood cells stained positively for peptide F (Bush et al.under review). The secondary antibody showedessentially no binding in the absence of the primaryantibody, suggesting that antibody binding wasspecific. Addition of peptide F to the cells did notgreatly change the percentage of cells that boundthe antibody, suggesting near endogenous satura-tion of receptors. However, normal rabbit serumused in place of the primary antibody also producedsimilar results. It was unlikely that binding to Fc-receptors occurred because cells were preincubated

with goat serum, human serum and calf serum as Fc-blocking agents. We were unable to reduce the bind-ing of normal rabbit serum to levels acceptable todetermine specific binding to peptide F (Bush et al.under review). Further research on determining thereceptor for peptide F needs to be done.

Effect of exercise on adrenal medullaryproenkephalins

Peptide F has been shown to elevate during exercise(Kraemer et al. 1985a, 1985b, 1990a, 1991; Bush et al.1998, under review; Triplett-McBride et al. 1998).Three of the studies by Kraemer et al. (1985a, 1985b,1992) compared peptide F responses in endurance-trained and untrained men. The pattern of release of peptide F in untrained men in response to cycle ergometer exercise (8-min stages) at increasingintensities to Vo2peak (Kraemer et al. 1985a, 1985b)was marked by increases in peptide F concentra-tions with increasing exercise intensity, with a surgeapproximately 5 min after the cessation of exercise.Concentrations of peptide F were reduced, thoughnot back to resting levels, by 15 min after exercise.With trained men, peptide F elevated at approx-imately 50% Vo2peak, dropped as exercise intensityapproached 100% Vo2peak, and then surged again at5 min after the cessation of exercise. By 15 min afterthe cessation of exercise peptide F concentrations

Peptide F receptors

Immune cell

Peptide F molecule

Primary antibody Secondary antibody

FITC

Fig. 16.6 Indirect immunophenotyping technique for detection of a peptide F receptor. Primary antibody = rabbit anti-peptide F. Secondary antibody = goat anti-rabbit and fluorescein isothiocyanate (FITC) labeled. (From Bush et al.under review.)

Page 225: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 209

had dropped, though not to resting concentrations(Kraemer et al. 1985a, 1985b). The Triplett-McBrideet al. (1998) investigation was performed in womenand the results were in contrast to prior studies inmen. For example, although the exercise mode wasthe same (cycle ergometer), only the trained groupshowed any peptide F response to the exercise, witha peak at the highest exercise intensity of 80%. Theuntrained group showed no exercise response inpeptide F.

Peptide F is also not under conscious control as demonstrated by Kraemer et al. (1992) in an in-vestigation utilizing hypnosis and the responses of peptide F to exercise. This study also comparedtrained to untrained men performing cycle ergome-ter exercise at 25% and 50% Vo2peak. The hypnosiscondition involved the suggestion of performingcycle ergometer exercise at 50% and 75% Vo2peakwhile subjects were actually cycling at 25% and 50%Vo2peak. The investigators found no significant dif-ferences in peptide F concentrations between condi-tions (control versus hypnosis) or between rest andexercise, suggesting that the actual intensity of exer-cise was still too low to elicit a peptide F response,despite the suggestion of higher intensity. Otherstudies by Kraemer et al. (1987, 1988, 1990a, 1991)examined peptide F responses to exercise in healthymen, and demonstrated increases with exercise oflong enough duration and high enough intensity.

Two studies (Kraemer et al. 1988, 1991) also util-ized cycle ergometer exercise, although varyingexercise protocols were employed. The 1988 studyexamined peptide F responses to steady state(80–85% Vo2peak) exercise to exhaustion with andwithout caffeine and at varying altitudes and alti-tude exposures (sea level, acute altitude and chronicaltitude). The investigators found that peptide Fconcentrations were higher at sea level (mid- andpost-exercise) and lower at chronic altitude (post-exercise) after caffeine ingestion. The results alsoindicated that post-exercise peptide F concentra-tions were lower than sea level concentrations atboth acute and chronic altitude after caffeine inges-tion. The exercise responses of peptide F weremixed. At sea level, only the caffeine ingestion trialproduced significant differences from pre-exerciseconcentrations for the mid- and post-exercise meas-

urements. At acute altitude, the only significant dif-ference from pre-exercise peptide F concentrationsoccurred at the mid-exercise measurement in thecaffeine ingestion trial. For the chronic altitude con-dition, the only significant increase in peptide F concentrations occurred with no caffeine ingestionat the mid-exercise measurement. The authors attri-buted most of these differences to the time of alti-tude exposure (17 days).

A study (Kraemer et al. 1991) examined peptide Fand catecholamine responses to exhaustive exerciseon a computerized cycle ergometer at various per-centages (36%, 55%, 73% and 100%) of maximal leg power. The focus of the study was to comparepatterns of response of peptide F, epinephrine, lactate and norepinephrine. There was a significantincrease in peptide F concentration immediatelypost-exercise at the 36% exercise intensity level,which was the longest in duration (3.5 min). Therewas also a significant increase in epinephrine imme-diate post-exercise at both the 36% and 55% exerciseintensity levels and at 15 min following the 100%exercise intensity level. The results indicated thatthese peptide F and epinephrine frequently have an inverse relationship at the higher intensity levels.There were also significant increases in norepine-phrine immediately after each exercise intensity, 5 min after the low to moderate exercise intensit-ies, and 15 min at the lowest exercise intensity level,suggesting a differential exercise response patternthan the encephalin. Similar exercise response patterns were observed with whole blood lactatefollowing all the exercise intensity levels. Althoughthese relationships were not significant, the authorspoint out that these relationships do not support thetheory of co-secretion of these substances from thechromaffin cells of the adrenal medulla as differen-tial response patterns to exercise may exist.

Another study (Kraemer et al. 1987) examinedpeptide F responses to steady state (70% Vo2max)treadmill exercise of long duration (100 min) before,during and after heat acclimation. The investigatorsfound that higher concentrations of peptide F in theheat were due to a reduction in degradation to [Met]-enkephalin. The investigators also found no sig-nificant differences pre- to post-exercise or betweentest days (before, during and after acclimation), and

Page 226: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

210 chapter 16

proposed that these findings may be a result ofdegradation processes in the longer duration exer-cise. In another investigation, Kraemer et al. (1990a)found that healthy men who performed treadmillexercise of increasing intensity (7-min stages) exhib-ited a peptide F response similar to that of theuntrained men in the 1985 investigations (Kraemeret al. 1985a, 1985b). The main difference betweenthese two groups of men was that the peak in peptide F concentration was at the maximal exer-cise intensity for the healthy men, not 5 min intorecovery as was found in the 1985 investigationsusing untrained men. This may be explained by the difference in fitness level and in exercise mode;cycling (non-weight-bearing) versus treadmill(weight-bearing).

The modality of exercise (aerobic versus anaero-bic) utilized can change the response of peptide F.Kraemer et al. (1992) studied the response of plasmapeptide F to a high intensity cycle ergometer test in10 healthy, active men. Four different intensitieswere utilized: 100% maximal leg power (equivalentto 318% Vo2max for 6 s), 73% maximal leg power(equivalent to 230% Vo2max for 15 s), 55% maximalleg power (equivalent to 175% Vo2max for 45 s) and36% maximal leg power (equivalent to 115% Vo2maxfor 180 s). This study showed the differential patternof epinephrine and peptide F in response to theexercise stress (i.e. epinephrine increased whilepeptide F decreased). With the longer duration ofexercise, peptide F was elevated (i.e. 115% Vo2maxfor 180 s) during exercise and returned to the restinglevel by 5–15 min post-exercise. This study gave initial insight into the difference in peptide F con-centration in response to an anaerobic-type exer-cise, such as high-intense short-duration cycling orresistance exercise. Such data demonstrated that theduration and/or volume of exercise may influencethe concentration of peptide F in the plasma.

Resistance exercise is anaerobic in nature. Twostudies have examined the responses of peptide Fconcentrations to resistance exercise (Bush et al.1998; Fry et al. 1998). Fry et al. (1998) examined thepeptide F response to an overtraining protocol.Subjects were tested following a high intensity resistance exercise training protocol (overtrained)versus a low intensity resistance exercise training

protocol (control). A high intensity training protocolwas performed for 2 weeks (i.e. 10 repetitions of 1-repetition maximum [1-RM] squat exercise everyday for 2 weeks) and a low intensity training proto-col (i.e. 50% 1-RM squats 1 day per week for 2 weeksand 1-RM testing 1 day per week for 2 weeks). Acutetesting was administered at the beginning, middleand end of the 2-week training period. Subjects performed continuous repetitions at 70% 1-RM untilexhaustion. No change in plasma peptide F concen-tration was observed in response to acute exercisestress and/or the overtraining protocol. The over-training protocol may have produced adaptationsin the adrenal medullary chromaffin system. Thelack of peptide F response to this type of trainingstimulus may have been due to inhibitory factorsnegatively affecting the proenkephalin biosynthesisinto the chromaffin cells (Wilson et al. 1982; Livett,B.G. 1984; Wilson 1991) or exocytotic suppression ofexisting peptide F from the chromaffin cells.

Reductions in the immune function with over-training have been observed (Mackinnon 1992).Such types of high intense training had negativeeffects on health and the immune system, since hormones such as cortisol and epinephrine, exhib-iting immunosuppressive effects, were elevated during this type of training (Kuipers & Keizer 1988;Fry et al. 1991, 1994). The lack of change in plasmapeptide F concentration in response to overtrainingmay have negatively impacted the immune system;and under such conditions plasma peptide F wouldnot be available to reverse any negative effects onimmune function produced by either cortisol orepinephrine (Kuipers & Keizer 1988; Fry et al. 1991,1994).

A second study (Bush et al. 1998) utilized a 16-setresistance exercise protocol. Four sets each of 10-RMand 15-RM bench press, bent over row, militarypress and squat exercise were performed by resist-ance trained men. The 10- and 15-RM protocols bothproduced a decrease in plasma peptide F concen-tration immediately following the acute resistanceexercise stress. At 15 min post-exercise, an increasein plasma peptide F was observed following onlythe 10-RM protocol. The higher forces involved inperforming the 10-RM protocol may account for thedifferential response in plasma peptide F. An 80%

Page 227: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 211

increase in peptide F concentration was observed 4 h post-exercise following both protocols. Thisstudy suggested that circulating peptide F may playa role during recovery periods from moderate resist-ance exercise to potentially increase immune func-tion during repair of disrupted muscle (Fridèn et al. 1983; McCully & Faulkner 1985; Round et al. 1987;Fielding et al. 1991; Nieman et al. 1995; McBride et al.1998). The increase in peptide F 4 h after resistanceexercise was not observed in the control group. Thisfinding was quite surprising. It may be speculatedthat there was an increase in production and/orsecretion of peptide F from the adrenal medulla, orthat peptide F bound to receptors on target tissueswere released.

Studies during heat acclimation in adults haveshown a reduction in plasma [Met]-enkephalin(Kraemer et al. 1987), potentially due to the increaseddegradation of the peptide in the circulatory systemof a decrease in the processing from the large pre-cursor unit. Circulating encephalin and opiatesreleased during stress (Viveros & Wilson 1983) andexercise (Howlett et al. 1984; Farrell et al. 1987) couldmediate the effects of stress reduction. There appearsto be a significant difference in the enzyme concen-trations responsible for hydrolyzing enkephalinssuch that the enkephalin concentrations were higherin a trained individual ( Jaskowski et al. 1989).

Exercise studies performed to exhaustion indicatean increase in plasma levels of [Met]-enkephalinand that this is observed in an exercise-intensitydependent manner (Sommers et al. 1990). It isknown that [Met]-enkephalin is co-released withepinephrine from the adrenal gland (Viveros et al.1979; Wilson et al. 1982; Wilson 1991). In an exer-cise study of moderate (70% Vo2max) to exhaustive exercise (120% Vo2max) focusing on the co-releasepatterns of epinephrine and [Met]-enkephalin, peakplasma epinephrine levels were observed 1 minpost-exercise, whereas peak plasma [Met]-enkepha-lin levels were observed during the moderate exer-cise intensity, declined during the high exerciseintensity and returned to baseline within 1 min post-exercise (Boone et al. 1992). Similar to the peptide Ffragment, a differential exercise response patternexists for epinephrine and [Met]-enkephalin.

Overtraining is defined as an increase in training

volume and/or intensity resulting in a decrease inphysical performance (Fry et al. 1991). An over-training study done by Fry et al. (1998) examinedresistance-trained adult men in which they per-formed 100% 1-RM barbell squats for a 2-weekperiod, resulting in a decrease in their 1-RM strengthin the overtrained state. Plasma testosterone andcortisol were decreased while in the overtrainedstate. Growth hormone, however, was not influ-enced by the high intensity resistance overtrainingprotocol. Epinephrine is elevated in response tohigh intensity resistance overtraining protocols (Fryet al. 1994) with no effect of overtraining response tocirculating levels of peptide F (Fry et al. 1998). Thisindicates the high intensity resistance overtrainingprotocol appeared to overwhelm the capacity of the adrenal medullary chromaffin cells to secretepeptide F.

Effect of exercise on peptide F in otherblood biocompartments

It has been reported that upon activation of lym-phocytes, a secretagogue was the proenkephalinprecursor (Roth et al. 1989). Lymphocytes differ intheir origination and maturation. The lymphocytesare a subclass of leukocytes, consisting of T- and B-cells, ranging in size from 6–10 µm and comprising20–25% of circulating leukocytes. Their functionsare crucial during an immune response and includerecognition of antigens, production of antibodiesagainst invading antigens, production of lympho-kines, action of cytotoxicity and memorization ofprevious antigen encounters. T-lymphocytes aregenerated within the bone marrow and mature inthe thymus, while B-lymphocytes are generated andmature within the bone marrow and fetal liver.Proliferation of mature lymphocytes occurs whilethey are surveying the system in the secondary lymphoid organs.

Approximately 1–2% of total lymphocytes recir-culate hourly, providing frequent communicationwith any foreign cells present in the system. Duringthis time of circulation, proenkephalin fragments(e.g. peptide F) may interact with these immunecells. T-lymphocytes are categorized by their func-tional ability: T-helper and T-cytotoxic cells. T- and

Page 228: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

212 chapter 16

B-lymphocytes are further differentiated by theirfunction during humoral-mediated or cellular-mediated immunity. Cell-mediated immunity dealswith cell-to-cell interactions and involves primarilythe attacking of foreign substances or antigens.Humoral-mediated immunity functions to supplythe system with antibodies (i.e. immunoglobulins)which have the ability to recognize the antigens(Sigal & Ron 1994). It is important to understand the difference between the two immune responsesas Hiddinga et al. (1994) reported. Specifically, asignificant cell-mediated response was producedbetween proenkephalin peptide fragments and T-cells, which then caused a humoral-mediated res-ponse (e.g. increased number of antibody-formingB-cells).

Based on the known interaction between peptideF and immune cells (Hiddinga et al. 1994), a studywas designed to examine the presence of peptide F within the white blood cell biocompartment ofblood. A progressive exercise stress protocol hadbeen shown to cause an increase in plasma peptide Fconcentration (Kraemer et al. 1985b, 1991). In a pro-gressive endurance exercise protocol, eight healthymen (21.0 ± 1.0 years) performed a 30-min cycleexercise at 80% Vo2max and returned for a secondsession under quiet control conditions. Blood sam-ples were taken pre-exercise, mid-exercise, immedi-ately post-exercise, 5 min, 15 min, 30 min and 60min post-exercise. Similar to previous research,there was an exercise-induced response of peptide F in the plasma biocompartment at 5 min post-exercise (Kraemer et al. 1985a, 1985b). Unique to this

pilot study was the presence of peptide F observedwithin the white blood cell biocompartment bothduring rest and following exercise. Subsequently,there was a decrease (p ≤ 0.05) in white blood cellbiocompartment peptide F concentration at 30 minpost-exercise (Fig. 16.7). It might be speculated that during early recovery periods from exercise(i.e. 5–15 min post-exercise), peptide F was bound to immune cell receptors and then internalized, andthus a response of peptide F to exercise may nothave been readily apparent until 30 min post-exercise. The presence of peptide F 30 min post-exercise in the white blood cell biocompartment wasobserved and may be attributed to a saturation ofpeptide F immune receptors.

The peptide F concentration observed in theplasma biocompartment following exercise was notsimilar to the concentration of peptide F observed inthe white blood cell biocompartment. It may be thatthere was a shift of peptide F concentration from the white blood cell biocompartment to the plasmabiocompartment, or that immune cell receptors forpeptide F were fully saturated following exercise(i.e. immediate, 5 min and 15 min post-exercise).These results indicated that a possible interactionbetween peptide F and cells of the immune systemexisted, and that exercise modulated the concen-tration of peptide F within this biocompartment.This initial pilot data was studied within the con-text of utilizing a progressive endurance exercisemodel. Examining the effect of acute resistance exercise on the peptide F concentration has been a topic of interest. Such data in this pilot study

Pep

tid

e F

(pm

ol·m

L–1)

0.12

0.10

0.08

0.06

0.04

0.02

0Pre-

exerciseMid-

exercise0 5 15

Post-exercise time (minutes)30 60

A A

A

A

A

Plasma WBC

Fig. 16.7 Peptide F concentrations in plasma and white blood cellbiocompartments at rest, mid-exercise and for 60 min into recoveryfollowing 30 min of cycling at 80%Vo2max. A = p ≤ 0.05 difference from corresponding pre-exercise,mean ± SE. WBC, white blood cellcompartment.

Page 229: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 213

supported the examination of peptide F within thewhite blood cell biocompartment, and the responseof peptide F in this biocompartment to resistanceexercise to secrete cytokines which produce notonly paracrine effects but also autocrine effects onthe same activated immune cells to up-regulatereceptor expression for further stimulation (Sigal & Ron 1994). Although the receptor for peptide Fhas not been characterized, it may potentially bindto cells within the blood biocompartments (i.e.immune cells). Erythrocytes possess peptide recep-tors for other hormones, such as insulin and insulin-like growth factor I (Horuk et al. 1993; Hagino et al.1994). There is also the possibility of peptide F in the red blood cell biocompartment interacting withsimilar peptide receptors or a yet to be determinedopioid.

In another study (Bush et al. under review) menaged 22 ± 0.9 years performed an acute resistanceexercise protocol where they completed six sets of10-RM squats on the Smith-squat machine. Bloodsamples for analysis of peptide F concentrationswere obtained at baseline, immediate post- and 15min post-exercise. There was a hierarchy of peptideF concentrations where the plasma biocompartmentshowed the highest (p < 0.05) concentration of pep-tide F versus the white blood cell and red blood cellbiocompartments at baseline and during recovery(Fig. 16.8). Furthermore, the concentration in thewhite blood cell biocompartment was higher thanthat of the red blood cell biocompartment (Fig. 16.8).This hierarchy of concentration was consistent both

under resting conditions and in response to theresistance exercise.

The increase in peptide F concentration in thewhite blood cell biocompartment occurred with asignificant increase in the total number of whiteblood cells measured in that biocompartment.During the recovery period, the number of peptideF molecules per white blood cell was significantlyincreased by nearly 50%, indicating the potentialsignificance of the interaction of peptide F withinthe white blood cell biocompartment. The inter-action with the white blood cell biocompartmentcan be supported by the fact that peptide F may bebinding to immune cell surface recpetors (Sibinga & Goldstein 1988), subsequently becoming inter-nalized, and resulting in less measurable peptide F in either the plasma or white blood cell biocom-partment. The increase in the number of peptide Fmolecules associated with immune cells duringrecovery can be potentially explained by: (i) adown-regulation of immune cell surface receptors;or (ii) an increase in production and secretion ofpeotide F from the adnreal medulla or immunecells, as observed by an increase in peptide F con-centrations with the white blood cell biocompart-ment. The measurable amount of peptide F in thered blood cell biocompartment may be reflective ofa shift in the peptide F among the three differentblood biocompartments. However, the physiolo-gical significance of the observance of peptide F inthe white and red blood cell biocompartmentsremains to be elucidated.

Pep

tid

e F

(pm

ol·m

L–1)

0.40

0.35

0.30

0.25

0.20

0.15

0.10

0.05

0Pre-exercise 0

Post-exercise time (minutes)15

A AB A AB

AAB

Plasma WBC RBC

Fig. 16.8 Comparison of peptide F concentration in threebiocompartments followingresistance exercise. A = p ≤ 0.05difference from correspondingplasma biocompartment; B = p ≤ 0.05difference from corresponding whiteblood cell biocompartment, mean ± SE. RBC, red blood cellbiocompartment; WBC, white bloodcell biocompartment.

Page 230: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

214 chapter 16

Summary

Neural stimulation is the primary mechanisminvolved with the activation and release of com-pounds in the adrenal medulla. Peptide F is an ECPsecreted from the chromaffin cells of the adrenalmedulla. Peptide F has been shown to be secretedalong with epinephrine during exercise (Kraemer et al. 1985a, 1985b, 1987, 1988, 1990a, 1991, 1992;Bush et al. 1998, under review; Triplett-McBride1998). However, the pattern of release of these twosubstances during and after exercise may differ(Kraemer et al. 1985b, 1990a, 1991). Interestingly, thepattern of release of peptide F during and after anacute bout of exercise also differs between highlytrained male endurance athletes and untrained men(Kraemer et al. 1985a, 1985b). Many of the biolo-gical functions of peptide F are unknown, but it hasbeen demonstrated that peptide F may improve theactivation and function of the T-cells of the immunesystem (Hiddinga et al. 1994). Peptide F has beenshown to enhance helper T-cell activation in vitroand subsequently cause increased production of

antibody by B-cells (Hiddinga et al. 1994). However,this relationship has not yet been studied in vivo.The complexity and degree of interactions betweenthe endocrine and immune systems are just begin-ning to be understood. Some investigators haveexamined the possibility of bi-directional regulationbetween these systems (Stein et al. 1985; Bateman et al. 1989). It has been shown that T-cells havereceptors for enkephalins (Wybran et al. 1979) andappear to have receptors specific for peptide F(Hiddinga et al. 1994). It has also been demonstratedthat T-cells can synthesize and release the smallECPs for autoregulation of function (Blalock 1992,1994). Therefore, it is possible that one of the mainroles of peptide F is to enhance immune cell func-tion to counteract and balance the suppressiveeffects of other endocrine hormones such as cortisoland epinephrine. While the modulation of endogen-ous peptide F may promote movement to varioustarget tissues, it appears that the mechanisms arerelated to adrenal medullary function, primarilyregulation of peptide F concentrations related to theresponse of stress (i.e. exercise).

ReferencesBastiaensen, E., Miserez, B. & De Potter, W.

(1988) Subcellular fractionation ofbovine ganglion stellatum: co-storage of noradrenaline, Met-enkephalin andneuropeptide Y in large ‘dense-cored’vesicles. Brain Research 442(1), 124–130.

Bateman, A., Singh, A., Kral, T. & Solomon,S. (1989) The immune–hypothalamic–pituitary–adrenal axis. Endocrine Reviews10, 92–112.

Blalock, J.E. (1992) Production of peptidehormones and neurotransmitters by theimmune system. Chemical Immunology52, 1–24.

Blalock, J.E. (1994) The syntax of immune–neuroendocrine communication.Immunology Today 15(11), 504–511.

Boarder, M.R. & McArdle, W. (1986)Breakdown of small enkephalinderivatives and adrenal peptide E byhuman plasma. Biochemical Pharmacology35, 1043–1047.

Boone, J.B., Sherraden, T., Pierzchala, K.,Berger, R. & Van Loon, G.R. (1992)Plasma Met-enkephalin andcatecholamine response to intenseexercise in humans. Journal of AppliedPhysiology 73(1), 388–392.

Brantl, V., Gramsch, C., Lottspeich, F. et al. (1986) Novel opioid peptidesderived from hemoglobin: hemorphins.European Journal of Pharmacology 125,309–310.

Brooks, S., Burrin, J., Cheetham, M.E. et al. (1988) The responses of thecatecholamines and β-endorphin to briefmaximal exercise in man. EuropeanJournal of Applied Physiology 57, 230–234.

Bush, J.A., Kraemer, W.J., Mastro, A.M. et al. (1998) Exercise and recoveryresponses of adrenal medullaryneurohormones to heavy resistanceexercise. Medicine and Science in Sportsand Exercise 31(4), 554–559.

Bush, J.A., Kraemer, W.J. & Mastro, A.M. (under review) Peptide Fimmunoreactivity in differentcirculatory biocompartments afterexercise stress (Peptides, in review).

Coupland, R.E. (1965) The Natural Historyof the Chromaffin Cell. Longman, London.

Coupland, R.E. (1972) The chromaffinsystem. In: Catecholamines: Handbook ofExperimental Pharmacology, vol. 33(Blaschko, H. & Muscholl, E. eds.).Springer-Verlag, Berlin: 16–39.

De Potter, W.P., Dillen, L., Annaert, W. et al. (1988) Evidence for the co-storageand co-release of neuropeptide Y andnoradrenaline from large dense coredvesicles in sympathetic nerves of thebovine vas deferens. Synapse 2(2),157–162.

Farrell, P.A., Gustafson, A.B., Morgan,W.P. & Pert, C.B. (1987) Enkephalin,catecholamines, and psychologicalmode alterations: effect of prolongedexercise. Medicine and Science in Sportsand Exercise 19, 347–353.

Fielding, R.A., Meredith, C.A., O’Reilly,K.P. et al. (1991) Enhanced proteinbreakdown after eccentric exercise inyoung and old men. Journal of AppliedPhysiology 71, 674–679.

Fridèn, J., Sjoström, M. & Ekblom, B. (1983)Myofibrillar damage following intenseeccentric exercise in man. InternationalJournal of Sports Medicine 4(3), 170–176.

Fry, A.C., Morton, A.R. & Keast, D. (1991)Overtraining in athletes, an update.Sports Medicine 12, 32–65.

Fry, A.C., Kraemer, W.J., Van Borselen, F.et al. (1994) Catecholamine responses toshort-term high-intensity resistance

Page 231: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

the adrenal medulla 215

exercise overtraining. Journal of AppliedPhysiology 77, 941–946.

Fry, A.C., Kraemer, W.J. & Ramsey, L.T.(1998) Pituitary–adrenal–gonadalresponses to high-intensity resistanceexercise overtraining. Journal of AppliedPhysiology 85(6), 2352–2359.

Genuth, S.M. (1988) The adrenal glands. In: Physiology (Berne, R.M. & Levy, M.N.,eds). C.V. Mosby Co., St. Louis, MI:950–982.

Hagino, H., Shii, K., Yokono, K. et al. (1994)Enzyme-linked immunosorbent assaymethod for human autophosphorylatedinsulin receptor: applicability to insulin-resistant states. Diabetes 43(2), 274–280.

Hansen, P.E. & Morgan, B.A. (1984)Structure-activity relationships inenkephalin peptides. In: The Peptides:Analysis, Synthesis, Biology (Udenfied, S.& Meienhofer, J. eds.). Academic Press,New York: 269–275.

Hiddinga, H.J., Katzenstein, G.E.,Middaugh, C.R. & Lewis, R.V. (1990)Secondary structure characteristics ofproenkephalin peptides E, B, and F.Neurochemical Research 15(4), 393–399.

Hiddinga, H.J., Isaak, D.D. & Lewis, R.V.(1994) Enkephalin-containing peptidesprocessed from proenkephalinsignificantly enhance the antibody-forming cell responses to antigens.Journal of Immunology 152, 3748–3759.

Horuk, R., Colby, T.J., Darbonne, W.C.,Schall, T.J. & Neote, K. (1993) Thehuman erythrocyte inflammatorypeptide (chemokine) receptor.Biochemical characterization,solubilization, and development of abinding assay for the soluble receptor.Biochemistry 32, 5733–5738.

Howlett, T.A., Tomlin, S., Ngahfoong, L. et al. (1984) Release of β-endorphin and met-enkephalin during exercise in normal women: response to training. British Medical Journal 288,1950–1952.

Hughes, J., Smith, T.W. Kosterlitz, H. et al.(1975) Identification of two relatedpentapeptides from the brain withpotent opiate agonist activity. Nature258, 577–579.

Ivanov, V.T., Karelin, A.A., Philippova,M.M., Nazimov, I.V. & Pletnev, V.Z.(1997) Hemoglobin as a source ofendogenous bioactive peptides: theconcept of tissue-specific peptide pool.Biopolymers 43, 171–188.

Jaskowski, M.A., Jackson, A.S., Raven, P.B.& Caffrey, J.L. (1989) Enkephalinmetabolism: effect of acute exercisestress and cardiovascular fitness.

responses during heat acclimation inhumans. Peptides 8, 715–719.

Kraemer, W.J., Rock, P.B., Fulco, C.S. et al.(1988) Influence of altitude and caffeineduring rest and exercise on plasmalevels of proenkephalin peptide F.Peptides 9, 1115–1119.

Kraemer, W.J., Diazdos, J.E., Gordon, S.E.et al. (1990a) The effects of gradedexercise on plasma proenkephalinpeptide F and catecholamine responsesat sea level. European Journal of Physiology61, 214–217.

Kraemer, W.J., Marchitelli, L., Gordon, S.E. et al. (1990b) Hormonal and growthfactor responses to heavy resistanceexercise protocols. Journal of AppliedPhysiology 69(4), 1442–1450.

Kraemer, W.J., Patton, J.F., Knuttgen, H.G.et al. (1991) Effects of high-intensity cycle exercise on sympathoadrenal–medullary response patterns. Journal ofApplied Physiology 70(1), 8–14.

Kraemer, W.J., Lewis, R.V., Triplett, N.T. et al. (1992) Effects of hypnosis onplasma proenkephalin peptide F and perceptual and cardiovascularresponses during submaximal exercise.European Journal of Applied Physiology 65,573–578.

Kuipers, H. & Keizer, H.A. (1988)Overtraining in elite athletes: reviewand directions for the future. SportsMedicine 6, 79–92.

Lewis, R.V. (1982) Enkephalin biosynthesisin the adrenal medulla. In: RegulatoryPeptides: from Molecular Biology toFunction, vol. 33 (Costa, E. & Trabucchi,M., eds.). Raven Press, New York:167–174.

Lewis, R.V. & Stern, A.S. (1983)Biosynthesis of the enkephalins andenkephalin-containing polypeptides.Annual Review of Pharmocology andToxicology 23, 353–372.

Lewis, R.V., Stern, A.S., Rossier, J., Stein, S.& Udenfriend, S. (1979) Putativeenkephalin precursors in bovine adrenal medulla. Biochemical andBiophysical Research Communications 89,822–829.

Livett, A.R., Dean, D.M., Whelan, L.G.,Udenfriend, S. & Rossier, J. (1981) Co-release of enkephalin andcatecholamines from cultured adrenalchromaffin cells. Nature 289, 317–319.

Livett, B.G. (1984) Adrenal medullarychromaffin cells in vitro. PhysiologicalReview 64, 1103–1161.

McBride, J.M., Kraemer, W.J., Triplett-McBride, N.T. & Sebastianelli, W. (1998)The effect of resistance exercise on free

Medicine and Science in Sports and Exercise21(2), 154–160.

Johnson, H.M., Smith, E.M., Torres, B.A. & Blalock, J.E. (1982) Regulation of the in vitro antibody response byneuroendocrine hormones. Proceedingsof the National Academy of Sciences 79,4171–4174.

Kaplan, N.R. (1988) The adrenal glands. In: Textbook of Endocrine Physiology(Griffin, J.E. & Ojeda, S.R., eds.). OxfordUniversity Press, New York: 245–272.

Katzenstein, G.E., Lund, D., Schultz, P. & Lewis, R.V. (1987) Target tissuedistribution of the proenkephalinpeptides F, E, and B. Biochemical andBiophysical Research Communications 146,1184–1190.

Kilpatrick, D.L., Lewis, R.V., Stein, S. & Udenfriend, S. (1980) Release ofenkephalins and enkephalin-containingpolypeptides from perfused beefadrenal glands. Proceedings of theNational Academy of Sciences USA 77(12),7473–7475.

Kilpatrick, D.L., Taniguchi, T., Jones, B.N.,Stern, A.S. & Shively, J.E. (1981) A highlypotent 3200 dalton adrenal opioidpeptide that contans both a [Met]- and[Leu]-enkephalin sequence. Proceedingsof the National Academy of Sciences of theUnited States of America 78, 3265–3268.

Kimura S., Lewis, R.V., Stern, A.S. et al. (1980) Probable precursors of[Leu]enkephalin and [Met]enkephalin in adrenal medulla: peptides of 3–5kilodaltons. Proceedings of the NationalAcademy of Sciences USA 77, 1681–1685.

Kjær, M. & Galbo, H. (1988) Effect ofphysical training on the capacity tosecrete epinephrine. Journal of AppliedPhysiology 64, 11–16.

Kjær, M., Christensen, N.J., Sonne, B.,Richter, E.A. & Galbo, H. (1985) Effect of exercise on epinephrine turnover intrained and untrained male subjects.Journal of Applied Physiology 59,1061–1067.

Kraemer, W.J., Noble, B., Culver, B. &Lewis, R.V. (1985a) Changes in plasmaproenkephalin peptide F andcatecholamine levels during gradedexercise in men. Proceedings of theNational Academy of Sciences of the UnitedStates of America 82, 6349–6351.

Kraemer, W.J., Noble, B., Robertson, K. &Lewis, R.V. (1985b) Response of plasmaproenkephalin peptide F to exercise.Peptides 6 (suppl. 2), 167–169.

Kraemer, W.J., Armstrong, L.E.,Marchitelli, L.J., Hubbard, R.W. & Leva, H. (1987) Plasma opioid peptide

Page 232: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

216 chapter 16

radical production. Medicine and Sciencein Sports and Exercise 30(1), 67–72.

McCarthy, D.A. & Dale, M.M. (1988) Theleukocytosis of exercise: a review andmodel. Sports Medicine 6, 333–363.

McCully, K.K. & Faulkner, J. (1985) Injuryto skeletal muscle fibers of micefollowing lengthening contraction.Journal of Applied Physiology 59, 119–121.

Mackinnon, L.T. (1992) Current Issues inExercise Science Series: Exercise andImmunology. Human Kinetics,Champaign, IL.

Marotti, T., Rabatic, S., Gabrilovac, J. (1993) A characterization of the in vivoimmunomodulation by Met-enkephalinin mice. International Journal ofImmunopharmacology 15(8), 919–926.

Martin, J., Prustowsky, M.B. & Angeletti,R.H. (1987) Preproenkephalin mRNA inT-cells, macrophages, and mast cells.Journal of Neuroscience Research 18, 82–87.

Nieman, D.C., Henson, D.A., Sampson,C.S. et al. (1995) The acute immuneresponse to exhaustive resistanceexercise. International Journal of SportsMedicine 16(5), 322–328.

Richter, G., Ballmann, M. & Conlon, J.M.(1986) Effect of streptozotocinadministration upon the serotonincontent of the pancreas and smallintestines of the rat. Hormone andMetabolic Research 18(10), 663–665.

Roth, K.A., Lorenz, R.G., Unanue, R.A. & Weaver, C.T. (1989) Nonopiate activeproenkephalin-derived peptides aresecreted by T helper cells. FASEB Journal3(12), 2401–2406.

Round, J.M., Jones, D.A. & Cambridge, G.(1987) Cellular infiltrates in humanskeletal muscle: exercise induceddamage as a model for inflammatorymuscle disease? Journal of theNeurological Sciences 82, 1–11.

catecholamins. Journal of AutonomicNervous System 7, 41–58.

Viveros, O.H., Diliberto, E.J., Hazum, E. & Chang, E.J. (1979) Opiate-likematerials in the adrenal medulla:evidence for storage and secretion withcatecholamines. Molecular Pharmacology16, 1101–1108.

Wasserman, K, Beaver, W. & Whipp, B.(1986) Mechanisms and pattern of bloodlactate increase during exercise in man.Medicine and Science in Sports and Exercise18, 344–352.

Wilson, S.P. (1991) Processing ofproenkephalin in adrenal chromaffincells. Journal of Neurochemistry 57(3),876–881.

Wilson, S.P., Chang, K.J. & Viveros, O.R.(1982) Proportional secretion of opioidpeptides and catecholamines fromadrenal chromaffin cells in culture.Journal of Neuroscience 2(8), 1150–1156.

Wybran, J., Appelbroom, T., Famaey, J.P. & Govaert, A. (1979) Suggestiveevidence for receptors for morphine and methionine-enkephalin on normalhuman blood T lymphocytes. Journal ofImmunology 123, 1068–1070.

Yanagihara, N., Oishi, Y., Yamamoto, H. et al. (1996) Phosphorylation ofchromogranin A and catecholaminesecretion stimulated by elevation ofintracellular Ca2+ in cultured bovineadrenal medullary cells. Journal ofBiological Chemistry 271(29), 17 463–17 468.

Zhao, Q., Garreau, I., Sannier, F. & Piot,J.M. (1997) Opioid peptides derivedfrom hemoglobin: hemorphins.Biopolymers 43(2), 75–98.

Zurawski, G., Benedik, M., Kamb, B.J. et al.(1986) Activation of mouse T-helper cellsinduces abundant preproenkephalinmRNA synthesis. Science 232, 772–775.

Schultzberg, M., Lundberg, J.M., Hökfelt,T. et al. (1978) Enkephalin-likeimmunoreactivity in gland cells andnerve terminals of the adrenal medulla.Neuroscience 3, 1169–1186.

Sibinga, N.E.S. & Goldstein, A. (1988)Opioid peptides and opioid receptors in cells of the immune system. AnnualReview of Immunology 6, 219–249.

Sigal, L.H. & Ron, Y. (1994) Cytokines. In:Immunology and Inflammation, BasicMechanisms, and Clinical Consequences(Sigal, L.H. & Ron, Y., eds). McGraw-Hill, New York: 482–490.

Simonds, W.F. (1988) The molecular basisof opioid receptor function. EndocrineReview 9, 200–212.

Sommers, D.K., Loots, J.M., Simpson, S.F.et al. (1990) Circulating met-enkephalinin trained athletes during rest,exhaustive treadmill exercise andmarathon running. European Journal ofClinical Pharmacology 38(4), 391–392.

Stein, M., Keller, S.E., Schleiter, S.J. (1985)Stress and immunomodulation: the roleof depression and neuroendocrinefunction. Journal of Immunology 135(2)(suppl.), 827–833.

Triplett-McBride, N.T., Mastro, A.M.,McBride, J.M. et al. (1998) Plasmaproenkephalin peptide F and human Bcell responses to exercise stress in fit and unfit women. Peptides 19(4),731–734.

Udenfriend, S. & Kilpatrick, D.L. (1984)Proenkephalin and the products of itsprocessing: chemistry and biology. In:The Peptides: Analysis, Synthesis, andBiology (Udenfriend, S. & Meienhofer, J.,eds), vol. 6. Academic Press, Orlando,FL: 25–67.

Viveros, O.H. & Wilson, S.P. (1983) Theadrenal chromaffin cell as a model tostudy the cosecretion of encephalin and

Page 233: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

217

The hypothalamic–pituitary–adrenal (HPA) axisconstitutes a hormonal network which is activatedduring stress. ‘Stress’ may take many forms, includ-ing acute episodes such as life-threatening illness,surgery or hemorrhage, or more chronic stressorssuch as depression or eating disorders. All of theseforms of stress have been extensively studied, andthe extent of the activation of the HPA axis quan-tified. Exercise, whether it is an acute episode ofstrenuous exercise or chronic endurance training isalso known to activate the HPA axis. This chapterwill briefly outline the physiological regulation ofthe HPA axis, followed by a comprehensive reviewof the specific effects of exercise and exercise train-ing on the axis in human subjects.

Overview of the regulation of cortisolproduction by the adrenal cortex

The target end-organ of the HPA axis, the adrenalcortex, secretes cortisol during basal conditions in a circadian pattern, with higher levels in the earlymorning hours which fall over the course of the day,reaching low levels by midnight (Krieger et al. 1971).Cortisol has a number of physiological roles. Theseinclude sodium and water balance and blood pres-sure control, maintenance of glucose homeostasis,adipogenesis, inhibition of osteoblast function andanti-inflammatory actions including suppression ofthe immune response (Stewart 2003). Cortisol isunder the influence of adrenocorticotropic hormone(ACTH) secreted by the anterior pituitary (Stewart2003). ACTH binds to corticotropin receptors in theadrenal cortex, which leads to cortisol production

and release (Catalano et al. 1986). Cortisol inhibits its own secretagogues, with a negative feedbackloop operating to inhibit ACTH from the pituitaryas well as acting at the hypothalamic level on bothcorticotropin-releasing hormone (CRH) and argi-nine vasopressin (AVP) (Keller-Wood & Dallman1984; Stewart 2003). This feedback tends to avoidprolonged and inappropriate periods of hypercor-tisolism. CRH and AVP, present in the parvicellularregion of the paraventricular nucleus in the hypo-thalamus (Pelletier et al. 1983), are the primary regu-lators of ACTH secretion (Orth 1992; Kjær, A. 1993).CRH is a 41-amino-acid peptide first isolated in thesheep in 1981 (Vale et al. 1981). It is a potent stimulusof both ACTH production and secretion (Orth 1992).CRH binds to specific high affinity CRH receptorson the corticotrope (Chen et al. 1993). Intracellularsignaling is via the protein kinase A/cyclic AMPsecond messenger system (Aguilera et al. 1983).Increased secretion of CRH is thought to play amajor role to increase ACTH and cortisol levels during many forms of acute stress (Chrousos 1992;Orth 1992). AVP, a 9-amino-acid peptide, binds to a specific receptors on corticotropes known as V3-(sometimes designated V1b) receptors (Sugimoto et al. 1994). In turn, the protein kinase C second mes-senger system is activated, leading to ACTH release(Liu et al. 1990). CRH and AVP are co-localized inthe median eminence of the hypothalamus (Whitnallet al. 1987). AVP acts synergistically with CRH to further amplify ACTH secretion during stress(Gillies et al. 1982, Rivier & Vale 1983). AVP is also acritical regulator of sodium and water balance and apotent vasoconstrictor (Jard 1988).

Chapter 17

Exercise and the Hypothalamic–Pituitary–Adrenal Axis

WARRICK J. INDER AND GARY A. WITTERT

Page 234: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

218 chapter 17

The presence of additional stimulatory and in-hibitory factors which may affect ACTH secretionhas been proposed (Grossman & Tsagarakis 1989;Alexander et al. 1996). A number of other hormones,cytokines and neurotransmitters interact with theHPA axis mainly through effects on CRH and lesscommonly AVP. There is evidence that leukemiainhibitory factor (LIF) is a stimulus to pituitaryACTH secretion (Auernhammer & Melmed 2000),but the physiological role of other factors interactingdirectly at the level of the pituitary or adrenalglands remains unclear. The regulation of the HPAaxis is summarized in Fig. 17.1.

The effect of acute exercise stress on the HPA axis and factors modulating the response

Mechanism of the cortisol response to exercise

Like other acute stressors, acute intense exercise is a potent activator of the HPA axis (Luger et al. 1987).The increase in plasma cortisol occurs in spite of anincrease in clearance from the circulation (Few1974). Even anticipation of competition may resultin a cortisol increase (Suay et al. 1999) and psycho-logical stress occurring prior to exercise increasesthe cortisol response (Kaciuba-Uscilko et al. 1994).

Studies in humans and large mammals have indic-ated an important role for both CRH and AVP in the exercise-induced stimulation of ACTH secretionwhich then causes the cortisol rise at the adrenallevel. Noting the limitations of peripheral CRHmeasurement, some (Elias et al. 1991; Harte et al.1995; Inder et al. 1998a) but not all (Luger et al. 1987;Wittert et al. 1991) studies have found an increase in plasma CRH after exercise (Fig. 17.2). Duration of the exercise may be important in explaining the differences, along with the sensitivity of the CRHassay. When exercise is performed under condi-tions of a continuous CRH infusion, at a level wherethe corticotrope CRH receptors would be fully saturated, a robust rise in ACTH and cortisol abovethe elevated baseline is noted (Smoak et al. 1991).This implies a factor additional to CRH being cru-cial in the ACTH response. In the pituitary venouseffluent of the exercising horse, plasma AVP levels

are markedly increased (Alexander et al. 1991). Inhumans, both acute high intensity exercise (Wittertet al. 1991) and prolonged submaximal exercise(Inder et al. 1998a) are associated with increases inplasma AVP that parallel ACTH and cortisol levels.The extent of the rise in AVP also appears to determine the degree to which the exercise-inducedactivation of the HPA axis is inhibited by exogenousglucocorticoids (Petrides et al. 1994). In a group ofexercising men pretreated with 4 mg dexametha-sone, four out of 11 were noted to exhibit a significantincrement in ACTH and cortisol following exercise.In this group, the plasma AVP response was sixtimes that of the group where dexamethasone abol-ished the ACTH/cortisol rise (Petrides et al. 1997).Subsequent studies comparing the group with-out dexamethasone suppressibility (designated highresponders) have also shown a greater integratedcortisol response to psychological stress (Singh et al.1999). This may identify a group of individuals who have a greater HPA axis response to differentstressors mediated via AVP. Changes in plasmaAVP are also correlated with alterations in plasmaosmolality, but the increase observed during intenseexercise is greater than expected due to osmolalitychanges alone (Wade & Claybaugh 1980). In par-ticular, the increase in plasma AVP correlates withchanges in plasma osmolality during prolonged sub-maximal exercise, but this relationship is lost dur-ing subsequent incremental exercise to exhaustion(Inder et al. 1998a). The reduction in plasma volumewhich occurs during exercise may also contribute tothe AVP response (Robertson & Athar 1976).

β-Endorphin is an opioid peptide derived fromproopiomelanocortin (POMC) (Morley 1981). Pre-viously it was thought to be secreted in equimolaramounts to ACTH, and a number of studies haveexamined the plasma β-endorphin response to exer-cise (Carr et al. 1981; Rahkila et al. 1987; Petraglia et al. 1988; Schwarz & Kindermann 1989). A majorproblem with much of this research is the find-ing of 100% cross-reactivity with β-lipotropin andacetylated forms of β-endorphin in most radioim-munoassays. Therefore, much of the β-endorphin-like immunoreactivity may not be opioid active.Using more specific immunoradiometric assays, it is now clear that in the basal state, β-endorphin is

Page 235: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 219

+

++

+

+

+

++

+

+

Processive(neurogenic) stress

(fear, restraint)

Systemic(physiological) stress(osmotic challenge,

macromolecules)

Systemic(physiological) stress(cytokines, hypoxia,

hemorrhage)

HippocampusPrefrontal cortex

Paraventricularnucleus

Raphe nucleus

NTS

Lateralseptum

Subfornical orgOVLT

MePO

HYPOTHALAMUS

BST HypothalamicGABAergic inputs

Hypothalamicglutamatergic inputs

CRHAVP

ACTH

PITUITARY

ADRENAL

Cortisol

Immunecells

IL-1, IL-2,IL-6, TNFα

toCNS

Arcuate,NPY and POMCneurons

Prostaglandins

Brain stemcatecholaminergic

inputs

Glucocorticoid receptors

Cytokine receptors

Leptin receptors

Hypothalamicglutamatergicinputs

5-HT

Brain stemvasculature

CRHneuron

GABAMeA

Fig. 17.1 Regulation of the hypothalamic–pituitary–adrenal (HPA) axis. Neuronal and neurotransmitter input tohypothalamic corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP) -containing neurons result inrelease of these hormones into the hypothalamic–hypophysial portal system. This causes an increase in secretion ofadrenocorticotropic hormone (ACTH) from the pituitary, which in turn stimulates cortisol release from the adrenal cortex.Cortisol acts at both pituitary and hypothalamic level via negative feedback to inhibit ACTH, CRH and AVP.

Page 236: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

220 chapter 17

undetectable in most normal subjects (Gibson et al.1993). True β-endorphin1–31 is released during exer-cise into peripheral blood in only about 50% of sub-jects, and represents only a small proportion ofβ-endorphin immunoreactivity (Harbach et al. 2000).It appears, however, that exercise does increase cent-ral levels of endogenous opioid peptides (Thoren et al. 1990). An infusion of naloxone, an opioidreceptor antagonist, increases the perceived effort ofexercise (Grossman et al. 1984; Sgherza et al. 2002).Highly trained athletes have evidence for increasedcentral opioid tone, probably induced by training(Inder et al. 1995). Basal plasma levels of β-endorphindo correlate with the subsequent ACTH response tonaloxone, a marker of central opioid tone (Inder etal. 1998b). Activation of endogenous opioid peptideshas been implicated in the positive mood effects of exercise. They have also been hypothesized in acausative role in exercise induced hypothalamicamenorrhea (Laatikainen 1991).

Effect of the intensity and duration of exercise

Short duration exercise to > 60% Vo2max results inACTH and cortisol release proportional to the inten-sity of the exercise (Davies & Few 1973; Howlett1987; Luger et al. 1987; Kjær, M. et al. 1988; Deuster et al. 1989; Wittert et al. 1991). Even high intens-ity exercise for as short as 1 min activates ACTH and cortisol secretion (Buono et al. 1986). Short-termsubmaximal exercise does not result in activation of the HPA axis even in conditions of extreme heat (Kenefick et al. 1998). Exercising at 50% Vo2maxfor 20 min does not cause an increase in cortisol levels, while at 70% Vo2max, ACTH and cortisol in-crease (Luger et al. 1987). When subjects have been

Co

rtis

ol (

nm

ol·L

–1)

1000

800

600

400

200

0(a)

ExerciseControl

†††† ††

******

**

AC

TH (

pm

ol·L

–1)

50

40

30

20

10

0(b)

**††

***††

***

††

††

***

AV

P (p

mo

l·L–1

)

10

8

6

4

2

0

(d)

CR

H (

pm

ol·L

–1)

5

4

3

2

1(c)

***††

***††

*

***††

–30 0 30

Time (min)

60 90

Fig. 17.2 (left) Changes in plasma levels (geometric mean± SEM) of (a) cortisol, (b) adrenocorticotropic hormone(ACTH), (c) corticotropin-releasing hormone (CRH) and(d) arginine vasopressin (AVP) in six male athletesundergoing a cycle ergometer-exercise protocol aiming tomaintain each athlete at 70% Vo2max for 1 h between 0 and60 min, followed by an incremental increase in absoluteintensity of 25 W every 2 min until exhaustion. Significantdifference from baseline (–30 min) *P < 0.05, **P < 0.01,***P < 0.001. Significant difference between exercise andcontrol time-matched data †P < 0.01, ††P < 0.001.

Page 237: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 221

subjected to a progressive increase in exercise intens-ity in 10-min blocks, commencing at 40% Vo2max, in-creased ACTH was only seen after 80% Vo2max wasreached (de Vries et al. 2000). Following 1 h of cycleergometer exercise at 70% Vo2max, cortisol levelsincreased from baseline; yet a further significantincrease in AVP, CRH, ACTH and cortisol wasobserved when the intensity was then progressivelyincreased over a 10-min period until exhaustion(Inder et al. 1998a). Using salivary as opposed toplasma cortisol, a cortisol rise following 1 h of exer-cise was observed only at 76% Vo2peak, but not at45% or 62%, while 40 min of exercise failed to in-crease salivary cortisol at any of the three intensities(Jacks et al. 2002).

These observations are best explained by the exer-cise being performed at or exceeding the anaerobicthreshold. Previous studies where the participantshave exercised at the individualized anaerobicthreshold have demonstrated that exercise belowthis point does not result in activation of the HPAaxis (Kindermann et al. 1982; Gabriel et al. 1992).During incremental exercise to exhaustion, plasmaACTH and β-endorphin rise only once the indi-vidual anaerobic threshold is reached (Schwarz &Kindermann 1990).

Although some studies have failed to demon-strate a cortisol rise in response to prolonged lowintensity exercise (Hoffman et al. 1994), ultra-longdistance running is associated with similar cortisolrises to repeated bursts of shorter, higher intensityexercise (Nagel et al. 1992). Cortisol levels at thecompletion of a 100-km ultramarathon were sig-nificantly elevated over baseline (Pestell et al. 1989).A 75-km cross-country ski race significantly elev-ated plasma cortisol levels (Vasankari et al. 1993). Ithas been suggested that the activation of the HPAaxis seen during prolonged low intensity exercise isdependent on the development of hypoglycemia(Tabata et al. 1991). In six subjects who exercised for14 h at 50% Vo2max, the cortisol, ACTH and CRHresponses were totally abolished when plasma glu-cose concentrations were maintained at pre-exerciselevels. The authors suggested a threshold plasmaglucose concentration of < 3.3 mmol·L–1 (Tabata et al. 1991). In an earlier study from the same group,cycling at 50% Vo2max for up to 3 h or exhaustion

elicited an increase in ACTH and cortisol only in thelatter part of the exercise when blood glucosedecreased (Tabata et al. 1990).

Timing of exercise

The HPA axis response to some stimuli may beinfluenced by the ambient basal cortisol level. Forexample, the cortisol increment may be less in themorning when basal levels are higher (DeCherneyet al. 1985). This is thought to be due negative feed-back inhibition. Kanaley et al. (2001) demonstratedthat although basal and peak cortisol levels inresponse to exercise were highest at 0700 h, theincremental response was greater compared to con-trol day when the exercise occurred at 2400 h. Incontrast, when comparing area under the curve andthe circadian baseline, no difference was found inwomen exercising at different times of the day(Thuma et al. 1995). When bouts of equivalent exer-cise are repeated later in the day, the ACTH and cortisol response to the second bout are greater thanthe first, or a single bout performed on another day (Ronsen et al. 2001a).

Type of exercise

Squats and intermittent higher intensity cyclingresult in a cortisol response in contrast to cyclingbeneath the anaerobic threshold which does not(Vanhelder et al. 1985). Estimation of the cortisolresponse to rowing has lead to conflicting results.Although a significant increases in plasma cortisolin response to maximal (7 min) and submaximal (40 min) rowing on rowing apparatus and 8 × 2000 mon the water (Snegovskaya & Viru 1993) has beenshown to occur in one study, a subsequent studywas unable to demonstrate an increase in plasmacortisol in response to maximal rowing to exhaus-tion on a rowing ergometer ( Jurimae & Jurimae2001). Similar results were found when the rowingwas performed at a lower intensity for 2 h, with nochange in plasma cortisol seen ( Jurimae et al. 2001).Cortisol levels were noted to increase followingkayak races of 19 and 42 km, with the extent of the rise being more pronounced in the longer race(Lutoslawska et al. 1991). Swimming for 30 min

Page 238: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

222 chapter 17

caused an increase in plasma cortisol at higherambient water temperatures but not at 20°C(Deligiannis et al. 1993). Activation of the HPA axiscan be induced by resistance exercise as well asthrough endurance exercise. During a high (100% ofsubject’s maximum) and moderate (70% of subject’smaximum) intensity strength workout, plasma cor-tisol increased more during the high intensity proto-col (Raastad et al. 2000). Three sets versus one set of resistance exercises resulted in a greater cortisolincrease (Gotshalk et al. 1997).

Age

In middle-aged men, resistance exercise consist-ing of sets of bench press, sit-ups and leg press,increased plasma cortisol concentrations (Häkkinen& Pakarinen 1995). This effect was not seen how-ever in women or elderly men. When comparingyoung, middle-aged and elderly men, Silvermanand Mazzeo (1996) found lower basal cortisol in theelderly sedentary subjects, but elevated basal levelsin the trained subjects of all ages. Peak cortisolresponse to maximal exercise showed an age-relateddecline, independent of training status. There wasno difference between men of different ages in thecortisol response to a 45-min submaximal exercise(Silverman & Mazzeo 1996).

Gender

Using 90 min of cycling at 80% of the anaerobicthreshold (approximately 50% Vo2max), there was no difference in cortisol response between men andwomen, matched for body mass index (BMI) andphysical fitness (Davis et al. 2000). Similar resultshave been obtained when 30 min of treadmill run-ning was employed (Kraemer, R.R. et al. 1989).Rahkila et al. (1987) found no difference betweenmen and women in cortisol and ACTH responseeither during a graded treadmill exercise to ex-haustion or anaerobic treadmill exercise. Therefore,both sexes appear to have a similar response to aero-bic endurance exercise. Following dexamethasone,however, the plasma cortisol and AVP response to high intensity (90–100% Vo2max) was greater in

women than men, possibly indicating a greater AVPresponse in women or a reduced sensitivity to glu-cocorticoid negative feedback (Deuster et al. 1998).African-American women may have an increasedACTH response to exercise, but cortisol responses aresimilar to Caucasian women (Yanovski et al. 2000).

Altitude

When comparative exercise has been done at lowand moderate altitude, cortisol levels were demon-strated to increase during exercise under both con-ditions; however, ACTH did not show an increaseabove baseline at moderate altitude (el-Migdadi et al. 1996). Comparison of athletes undergoinginterval training at sea level and 1800 m did notreveal any statistically significant difference in cor-tisol response, although the sympathetic nervoussystem response was greater at higher altitude(Niess et al. 2003). Marathon runners competing athigh altitude develop higher basal cortisol levels on acclimatization which increase further on com-pletion of the race (Marinelli et al. 1994). Similar data were observed in a group of healthy volunteersundertaking a rigorous trekking expedition in theHimalayas, where cortisol levels increased after 2 weeks (Martignoni et al. 1997). While circadianrhythms were maintained, 30% of the subjectsdemonstrated failure of dexamethasone suppres-sion (Martignoni et al. 1997). Overall, it appears thatacclimatization to altitude results in an increase inbasal cortisol levels compared to sea level, while thecortisol response to exercise is preserved.

Nutrition

Several studies have examined the effects of dietarychanges and supplement use, before, during andafter exercise, on the plasma cortisol response.Ingestion of carbohydrates during prolonged (2.5 h)cycling or running at approximately 70% Vo2maxresulted in a lower cortisol response to the exerciseand a lower rating of perceived exertion (Utter et al.1999). Similar results were obtained by Deuster et al.(1992) who observed that ingestion of a 7% glucosepolymer/fructose/electrolyte solution at a rate of

Page 239: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 223

200 mL every 30 min abolished the exercise-inducedcortisol rise during a 2-h run at 60–65% Vo2max com-pared to exercise ingesting an equal volume ofwater. Murray et al. (1991) using a carbohydratesolution compared to a water placebo, also noted animprovement in a 4.8-km performance test after 2 hof prolonged cycling exercise at 65–75% Vo2max, inaddition to a reduction in the ACTH and cortisolresponse.

Three days on a ketogenic diet of equal energycontent resulted in higher cortisol levels both beforeand after exercise in comparison to a control mixeddiet (Langfort et al. 1996). Glycerol, which has beensuggested as an aid to maintain hydration duringexercise, did not result in any difference in plasmacortisol response to 1 h of cycle ergometer exerciseat 70% Vo2max followed by an incremental increasein intensity to exhaustion (Inder et al. 1998c).

Creatine is a popular nutritional supplementamong athletes. Short-term creatine supplementa-tion for 5 days did not alter the cortisol response to a1 h session of heavy resistance training, althoughlevels tended to be higher during recovery follow-ing the creatine (Op’t Eijnde & Hespel 2001). Post-exercise feeding with whole food, supplementaldrink, carbohydrate beverage or placebo made nodifference to cortisol levels measured over 24 h fol-lowing exercise (Bloomer et al. 2000).

The effect of exercise training on theHPA axis

The objective of training is to optimize human per-formance; training produces a number of neuro-endocrine adaptations, which result in alterations of the activity of the HPA axis. This response isdetermined by the training volume, intensity, typeof exercise, optimum periods of rest (regeneration)and poorly defined psychological factors.

Effect of training on basal activity of the HPA axis

The nature of the effect of training on basal activityof the HPA axis remains unresolved. Normalizationof plasma cortisol levels after prolonged enduranceexercise may take up to 18–24 h (Lutoslawska et al.

1991; Heitkamp et al. 1996). The recovery fromintense, prolonged exercise in trained athletes isassociated with greater plasma ACTH levels, butsimilar plasma cortisol levels as compared to con-trol subjects (Duclos et al. 1997). Intensive trainingfor an ultramarathon (Tharp & Buuck 1974; Pestell et al. 1989; Wittert et al. 1996), or experimentally inrecreational athletes (Lehmann et al. 1993), has beenshown to increase pituitary ACTH secretion with-out affecting plasma cortisol levels. A number ofother studies are consistent with this. In male andfemale joggers, a season of training was withoutaffect on basal plasma cortisol levels (Ronkainen et al. 1986). In middle- and long-distance runners,neither an increase in training intensity nor 12weeks of intense training in competitive swimmers(Mujika et al. 1996) affected basal plasma cortisollevels (Lehmann et al. 1992; Flynn 1997). While thissuggests reduced adrenal responsiveness to ACTH,Duclos et al. (1998) showed that that responsivenessto ACTH did not appear to be decreased afterendurance training (Duclos et al. 1998) but the sens-itivity of the HPA axis to glucocorticoid feedback, at least at the level of the pituitary, was decreased(Duclos et al. 2001). In trained men, there is adecreased sensitivity of monocytes to cortisol 24 hafter the last bout of exercise. This may be related toa process of desensitization that may act to protectthe body from prolonged, exercise-induced cortisolsecretion (Duclos et al. 2003).

In contrast to these studies, it has been shown thatintensive training on a treadmill increased restingplasma cortisol levels without affecting basal ACTH(Kraemer, W.J. et al. 1989). In competitive swimmers,a short-term increase in training distance mayincrease basal plasma cortisol levels (without neces-sarily affecting performance) (Kirwan et al. 1988),and trained cyclists have been reported to havehigher basal cortisol levels than untrained controls(Silverman & Mazzeo 1996).

Age, gender, nutrition, mood, type and durationof activity and training status may all contribute tothe effects of physical training on the HPA axis.There is no difference in the response of male andfemale athletes to a sudden increase in training(O’Connor et al. 1991). In prepubescent gymnasts, 5

Page 240: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

224 chapter 17

consecutive days of training (3 h each day at mod-erate intensity), had no significant effect on basalplasma cortisol levels (Rich et al. 1992). In contrast,in a similar group of gymnasts 7–16 weeks of train-ing resulted in increased basal cortisol levels, butenergy intake was 31% below recommended levels(Filaire et al. 2003). Accordingly, the increased corti-sol levels were likely the consequence of an energydeficit rather than training per se. In well-nourishedyoung male gymnasts training had no significanteffect in basal cortisol levels (Daly et al. 1998).

The response of plasma cortisol may be deter-mined by both the duration and the nature of thetraining program, because sprint interval (asso-ciated with a substantial anaerobic component totraining) but not endurance training has beenobserved to increase basal plasma cortisol (Kraemer,W.J. et al. 1989; Wittert 1996). An increase in trainingvolume as opposed to intensity has been observedto induce a decrease in both resting and exercise-induced cortisol levels (Lehmann et al. 1992), whichis characteristic of an overtraining syndrome. Incontrast a twofold increase in training volume hasbeen shown not to affect plasma cortisol levels, andthere was no difference in the endocrine responsesto an increase in training volume with cross-trainingas compared to mode-specific training (Flynn et al.1997). The hormonal response to 5 weeks of aerobictraining is similar regardless of whether trainingwas carried out under conditions to simulate in-creased altitude (2500 m) or at sea level (Engred et al. 1994). Similarly, season appears to have noeffect on the response to exercise training (Ronsen et al. 2001b). In elderly men there is substantial indi-vidual variability in the effect of endurance trainingon the HPA axis, although in general the changesappear to be similar to those that occur in youngermen (Struder et al. 1999).

The effect of resistance exercise training on basalplasma cortisol levels is variable, and either nochange (Häkkinen et al. 1988a, 1988b; Fry et al. 1994)or a decrease has been reported (Alen et al. 1988); the difference probably related to lower trainingvolumes. Increasing resistance, training volume, orintensity may result in an increase in resting cortisollevels (Fry & Kraemer 1997). A doubling of trainingvolume has also been observed to result in a

decrease in intensity-dependent, exercise induced,increases in cortisol levels (Fry & Kraemer 1997).Although 2 years of intensive weight training waswithout affect on basal cortisol levels in adolescentmales (Fry et al. 1994), 1 week of an overreachingstimulus induced an increase in early morning corti-sol levels in this group of subjects (Fry et al. 1994). Inyoung men, high-intensity weight training sufficientto induce an overtrained state resulted in a slightlyincreased exercise-induced testosterone/cortisol ratiobut decreased exercise-induced cortisol. This hor-monal profile is distinctly different from what hasbeen previously reported for other types of over-training, indicating that high relative intensity resist-ance exercise overtraining may not be successfullymonitored via circulating testosterone and cortisol(Fry et al. 1998).

Effect of training on the response to subsequentacute exercise

The cortisol response to exercise of the same relativeintensity (Rolandi et al. 1985; Deuster et al. 1989) isnot affected by training, although the response tothe same absolute intensity may decrease (Buono et al. 1987; Deuster et al. 1989; Botticelli et al. 1992;Hickson et al. 1994). In other words, some degree of adaptation occurs. In contrast, activation of theHPA axis in response to supramaximal exercise is greater in endurance-trained subjects than inuntrained subjects (Furell et al. 1987; Snegovskaya &Viru 1993). The nature of the exercise training mayto some extent determine the response of the HPAaxis to subsequent acute exercise; when a substan-tial anaerobic component is present, the cortisolresponse to subsequent exercise may be increased(Fry et al. 1997).

Exercise and abnormalities of the HPA axis

Overreaching and overtraining

If adaptation to the increased training regimen doesnot occur, or if insufficient recovery time is includedin training regimens, overreaching and subsequentlyovertraining may occur. Overreaching may be con-

Page 241: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 225

sidered as short-term overtraining and may be anormal part of athletic training, or may be seen as ashort-term consequence of ultra-endurance events.By contrast overtraining results in a state describedas burnout or staleness, which is characterized byincreased fatigue, altered mood state, increasedinfections and suppressed reproductive function.Overtraining may be, at least in part, the conse-quence of inadequate periods of ‘regeneration’(Lehmann et al. 1997).

From the HPA axis standpoint, it has been sug-gested that the earliest stage of overreaching (orvery early overtraining) may be reflected by a re-duced responsiveness to ACTH, which is initiallycompensated for by an increased pituitary ACTHresponse but a decreased adrenal cortisol response(Wittert et al. 1996; Lehmann et al. 1997). When fullyevolved, the overtraining syndrome is charac-terized by both increased basal and 24-h urinarycortisol levels and a reduced ACTH and cortisolresponse to physical activity (Barron et al. 1985;Lehmann et al. 1992). In highly trained distance run-ners who undertook a 38% increase in trainingintensity over 3 weeks, six of the subjects developedsustained fatigue and the increase in serum cortisol,normally induced by 30 min of submaximal exer-cise, was lost (Verde et al. 1992). The severest andmost fully evolved form of the overtraining syn-drome is characterized by underactivity of both the HPA axis and sympathoadrenal system. Thiscomplete pattern is only observed subsequent tohigh-volume endurance overtraining at high caloricdemands (Figs 17.3 and 17.4) (Lehmann et al. 1998).

Whether overtraining induced by high volumeresistance exercise produces similar effects to highlyaerobic activities is not entirely clear. After maximal-intensity-resistance exercise training, basal plasma

cortisol or ACTH levels appear unaffected, but the responsiveness to exercise is decreased (Fry &Kraemer 1997). Some data suggests that hormoneresponses to exercise load are superior in indicatingheavy training-induced stress when compared withresting hormone levels. These responses indicateddecreased adrenocortical activity. However, sincemarked individual differences were found in train-ing- and overtraining-induced hormonal changes,individual hormonal profiles are needed to follow-up training effects (Uusitalo et al. 1998).

Athletic amenorrhea

Exercise-associated disorders of the reproductiveaxis in women are associated with abnormalities ofthe HPA axis; the response of plasma cortisol toboth maximal and submaximal acute exercise isreduced in amenorrheic compared to eumenorrheicathletes (Loucks & Horvath 1984; De Souza 1991).Amenorrheic athletes have been found to havehigher mean basal (Ding et al. 1988; Loucks et al.1989) early morning (Loucks et al. 1989) or mid-afternoon- (De Souza et al. 1991) blood and 24-hurine (Loucks et al. 1989) cortisol levels. There is alsoevidence that basal CRH stimulation is increased(Loucks et al. 1989; Hohtari et al. 1991) and thatadrenal sensitivity to ACTH is reduced (De Souza et al. 1991, 1994).

Summary

Trained and untrained individuals have a robustincrease in ACTH and cortisol in response to acutehigh intensity exercise. This is mediated via thehypothalamus by both CRH and AVP. The extent of the cortisol rise is proportional to the relative

Short-termintensified training

ACTH

Cortisol

Welladapted

Overreaching Overtraining

Fig. 17.3 Effect of exercise trainingon plasma adrenocorticotropichormone (ACTH) and cortisol levels.

Page 242: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

226 chapter 17

intensity of the exercise as expressed as a percent-age of Vo2max. Older people may have a bluntedresponse, while no difference exists between maleand female subjects. At exercise levels below theanaerobic threshold, the duration of exercise has to

be more prolonged to produce a significant increasein cortisol levels. The effect of resistance exercise onthe HPA axis appears to be most variable, and thereare both gender and age-specific affects. Variableresponses have been observed during other formsof exercise, such as swimming and rowing. Carbo-hydrate supplementation during more prolongedexercise can attenuate or abolish the cortisol rise,which may implicate relative hypoglycemia as a potential factor inducing the activation of the HPA axis in this setting. At altitude, exercise stillresults in a significant rise in plasma cortisol,although basal cortisol levels are higher followingacclimatization.

Although a great deal has been learned about theeffects of physical activity on the HPA axis, there isstill much conflicting information and many areasof confusion. The response of the HPA axis to any stress is dependent not only on the nature of the stress, but also on the environment in which thestress is imposed as well the inherent characteristicsof the individual concerned (genetic factors, gender,personality, prior experience), concomitant stres-sors and nutritional state. In addition, the timingand manner in which samples are collected will alsoinfluence the results obtained.

In general, it appears that the response to short-term intensified training is an increased plasma cortisol level, particularly if an anaerobic element isinvolved. Progressively adaptation occurs, with adecreasing adrenal response to ACTH to exercise atthe same relative intensity. With overreaching, theabsolute cortisol response is decreased, and whenovertraining occurs, HPA axis activity is decreasedas a whole (Barron et al. 1985). The precise factorsthat may modify an appropriate adaptive responseand lead to overreaching or overtraining remain tobe determined.

The extent to which abnormalities of the HPA axisare simply consequences of, or are inherent to thepathophysiology of, a variety of conditions relatedto exercise training are not entirely clear. Whetheractivity of the HPA axis can be used as a marker of training stress and adaptation, by establishing the activity of and following longitudinally the HPA axis of individual athletes remains to be determined.

Co

rtis

ol (

nm

ol·L

–1)

550500450400350300250200150100500

3 4 5 6 7 8 9 10Time (h)(a)

Time (h)(b)

11 12 13 14 15 16

ControlsAthletes

AC

TH (

ng

·L–1

)

9080706050403020100

3 4 5 6 7 8 9 10

Athletes(c)

Co

rtis

ol (

mm

ol·2

4h

–1)

250

200

150

100

50

0Controls

11 12 13 14 15 16

ControlsAthletes

Fig. 17.4 Adaptation of the hypothalamic–pituitary–adrenal (HPA) axis to chronic exercise stress in humans.Mean (± SEM) plasma concentrations of (a) cortisol, (b)adrenocorticotropic hormone (ACTH) and (c) a 24-hurinary free cortisol excretion in ultramarathon athletesand control subjects.

Page 243: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 227

References

Aguilera, G., Harwood, J.P., Wilson, J.X. et al. (1983) Mechanisms of action ofcorticotrophin-releasing factor and otherregulators of corticotropin release in rat pituitary cells. Journal of BiologicalChemistry 258, 8039–8045.

Alen, M., Pakarinen, A., Häkkinen, K. &Komi, P.V. (1988) Responses of serumandrogenic-anabolic and catabolichormones to prolonged strengthtraining. International Journal of SportsMedicine 9, 229–233.

Alexander, S.L., Irvine, C.H., Ellis, M.J. & Donald, R.A. (1991) The effect of acute exercise on the secretion ofcorticotropin-releasing factor, argininevasopressin, and adrenocorticotropin as measured in pituitary venous bloodfrom the horse. Endocrinology 128, 65–72.

Alexander, S.L., Irvine, C.H. & Donald,R.A. (1996) Dynamics of the regulationof the hypothalamo–pituitary–adrenal(HPA) axis determined using anonsurgical method for collectingpituitary venous blood from horses.Frontiers in Neuroendocrinology 17, 1–50.

Auernhammer, C.J. & Melmed, S. (2000) Leukemia-inhibitory factor-neuroimmune modulator of endocrinefunction. Endocrine Reviews 21, 313–345.

Barron, J.L., Noakes, T.I., Levy, W., Smith,C. & Millar, R.P. (1985) Hypothalamicdysfunction in overtrained athletes.Journal of Clinical Endocrinology andMetabolism 60, 803–806.

Bloomer, R.J., Sforzo, G.A. & Keller, B.A.(2000) Effects of meal form andcomposition on plasma testosterone,cortisol, and insulin following resistanceexercise. International Journal of SportNutrition Exercise and Metabolism 10,415–424.

Botticelli, G., Bacchi Modena, A., Bresciani,D. et al. (1992) Effect of naltrexonetreatment on the treadmill exercise-induced hormone release inamenorrheic women. Journal ofEndocrinological Investigation 15,839–847.

Buono, M.J., Yeager, J.E. & Hodgdon, J.A.(1986) Plasma adrenocorticotropin andcortisol responses to brief high-intensityexercise in humans. Journal of AppliedPhysiology 61, 1337–1339.

Buono, M.J., Yeager, J.E. & Sucec, A.A.(1987) Effect of aerobic training on theplasma ACTH response to exercise.Journal of Applied Physiology 63,2499–2501.

runners. Journal of Applied Physiology 76,1302–1309.

Deligiannis, A., Karamouzis, M., Kouidi,E., Mougios, V. & Kallaras, C. (1993)Plasma TSH, T3, T4 and cortisolresponses to swimming at varying watertemperatures. British Journal of SportsMedicine 27, 247–250.

Deuster, P.A., Chrousos, G.P., Luger, A. et al. (1989) Hormonal and metabolicresponses of untrained, moderatelytrained, and highly trained men to threeexercise intensities. Metabolism 38,141–148.

Deuster, P.A., Singh, A., Hofmann, A.,Moses, F.M. & Chrousos, G.C. (1992)Hormonal responses to ingesting wateror a carbohydrate beverage during a 2 hrun. Medicine and Science in Sports andExercise 24, 72–79.

Deuster, P.A., Petrides, J.S., Singh, A. et al. (1998) High intensity exercisepromotes escape of adrenocorticotropinand cortisol from suppression bydexamethasone: sexually dimorphicresponses. Journal of ClinicalEndocrinology and Metabolism 83,3332–3338.

Ding, J.H., Sheckter, C.B., Drinkwater, B.L.,Soules, M.R. & Bremner, W.J. (1988)High serum cortisol levels in exercise-associated amenorrhea. Annals ofInternal Medicine 108, 530–534.

Duclos, M., Corcuff, J.B., Rashedi, M.,Fougere, V. & Manier, G. (1997) Trainedversus untrained men: differentimmediate post-exercise responses ofpituitary adrenal axis. A preliminarystudy. European Journal of AppliedPhysiology 75, 343–350.

Duclos, M., Corcuff, J.B., Arsac, L. et al.(1998) Corticotroph axis sensitivity afterexercise in endurance-trained athletes.Clinical Endocrinology 48, 493–501.

Duclos, M., Corcuff, J.B., Pehourcq, F. &Tabarin, A. (2001) Decreased pituitarysensitivity to glucocorticoids inendurance-trained men. EuropeanJournal of Endocrinology 144, 363–368.

Duclos, M., Gouarne, C. & Bonnemaison,D. (2003) Acute and chronic effects of exercise on tissue sensitivity toglucocorticoids. Journal of AppliedPhysiology 94, 869–875.

Elias, A.N., Wilson, A.F., Pandian, M.R. et al. (1991) Corticotropin releasinghormone and gonadotropin secretion in physically active males after acuteexercise. European Journal of Applied

Carr, D.B., Bullen, B.A., Skrinar, G.S. et al.(1981) Physical conditioning facilitatesthe exercise-induced secretion of β-endorphin and β-lipotropin in women.New England Journal of Medicine 305,560–563.

Catalano, R.D., Stuve, L. & Ramachandran,J. (1986) Characterization ofcorticotropin receptors in humanadrenocortical cells. Journal of ClinicalEndocrinology and Metabolism 62,300–304.

Chen, R., Lewis, K.A., Perrin, M.H. & Vale,W.W. (1993) Expression cloning of ahuman corticotropin-releasing-factorreceptor. Proceedings of the NationalAcademy of Science of the United States ofAmerica 90, 8967–8971.

Chrousos, G.P. (1992) Regulation anddysregulation of the hypothalamic–pituitary–adrenal axis. Thecorticotropin-releasing hormoneperspective. Endocrinology andMetabolism Clinics of North America 21,833–858.

Daly, R.M., Rich, P.A. & Klein, R. (1998)Hormonal responses to physical trainingin high-level peripubertal malegymnasts. European Journal of AppliedPhysiology and Occupational Physiology79, 74–81.

Davies, C.T. & Few, J.D. (1973) Effects ofexercise on adrenocortical function.Journal of Applied Physiology 35, 887–891.

Davis, S.N., Galassetti, P., Wasserman,D.H. & Tate, D. (2000) Effects of genderon neuroendocrine and metaboliccounterregulatory responses to exercisein normal man. Journal of ClinicalEndocrinology and Metabolism 85,224–230.

DeCherney, G.S., DeBold, C.R., Jackson,R.V. et al. (1985) Diurnal variation in theresponse of plasma adrenocorticotropinand cortisol to intravenous ovinecorticotropin-releasing hormone. Journalof Clinical Endocrinology and Metabolism61, 273–279.

De Souza, M.J., Maguire, M.S., Maresh,C.M. et al. (1991) Adrenal activation andthe prolactin response to exercise ineumenorrheic and amenorrheic runners.Journal of Applied Physiology 70,2378–2387.

De Souza, M.J., Luciano, A.-A., Arca, J.C.,Demers, L.M. & Loucks, A.B. (1994)Clinical tests explain blunted cortisolresponsiveness but not mildhypercortisolism in amenorrheic

Page 244: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

228 chapter 17

Physiology and Occupational Physiology62, 171–174.

Engfred, K., Kjær, M., Secher, N.H. et al.(1994) Hypoxia and training inducedadaptation of hormonal responses toexercise in humans. European Journal ofApplied Physiology 68, 303–309.

Few, J.D. (1974) Effect of exercise on thesecretion and metabolism of cortisol inman. Journal of Endocrinology 62, 341–353.

Filaire, E., Jouanel, P., Colombier, M.,Begue, R.J. & Lac G. (2003) Effects of 16 weeks of training prior to a majorcompetition on hormonal andbiochemical parameters in young elite gymnasts. Journal of PediatricEndocrinology and Metabolism 16, 741–750.

Flynn, M.G., Pizza, F.X. & Brolinson, P.G.(1997) Hormonal responses to excessivetraining: influence of cross training.International Journal of Sports Medicine 18,191–196.

Fry, A.C., & Kramer, W.J. (1997) Resistanceexercise overtraining and overreaching.Neuroendocrine responses. SportsMedicine (Auckland, NZ) 23, 106–129.

Fry, A.C., Kraemer, W.J., Stone, M.H. et al. (1994) Endocrine responses tooverreaching before and after 1 year ofweightlifting. Canadian Journal of AppliedPhysiology 19, 400–410.

Fry, A.C., Kraemer, W.J. & Ramsey, L.T.(1998) Pituitary–adrenal–gonadalresponses to high-intensity resistanceexercise overtraining. Journal of AppliedPhysiology 85, 2352–2359.

Furell, P.A., Kjær, M., Bach, F.W. & Galbo, H. (1987) Beta-endorphin andadrenocorticotropin response tosupramaximal treadmill exercise intrained and untrained males. ActaPhysiologica Scandinavica 130, 619–625.

Gabriel, H., Schwarz, L., Steffens, G. & Kindermann, W. (1992)Immunoregulatory hormones,circulating leucocyte and lymphocytesubpopulations before and afterendurance exercise of differentintensities. International Journal of SportsMedicine 13, 359–366.

Gibson, S., Crosby, S.R. & White, A. (1993)Discrimination between β-endorphinand β-lipotrophin in human plasmausing two-site immunoradiometricassays. Clinical Endocrinology 39,445–453.

Gillies, G.E., Linton, E.A. & Lowry, P.J.(1982) Corticotropin releasing activity ofthe new CRF is potentiated several timesby vasopressin. Nature 299, 355–357.

Gotshalk, L.A., Loebel, C.C., Nindl, B.C. et al. (1997) Hormonal responses of

Hohtari, H., Salminen Lappalainen, K. & Laatikainen, T. (1991) Response ofplasma endorphins, corticotropin,cortisol, and luteinizing hormone in the corticotropin-releasing hormonestimulation test in eumenorrheic andamenorrheic athletes. Fertility andSterility 55, 276–280.

Howlett, T.A. (1987) Hormonal responsesto exercise and training: a short review.Clinical Endocrinology 26, 723–742.

Inder, W.J., Hellemans, J., Ellis, M.J. et al. (1995) Elevated basaladrenocorticotropin and evidence forincreased central opioid tone in highlytrained male athletes. Journal of ClinicalEndocrinology and Metabolism 80,244–248.

Inder, W.J., Hellemans, J., Swanney, M.P.,Prickett, T.C. & Donald, R.A. (1998a)Prolonged exercise increases peripheralplasma ACTH, CRH, and AVP in maleathletes. Journal of Applied Physiology 85,835– 841.

Inder, W.J., Livesey, J.H. & Donald, R.A.(1998b) Peripheral plasma levels of β-endorphin in alcoholics and highlytrained athletes and the relationship to a measure of central opioid tone.Hormone and Metabolic Research 30,523–525.

Inder, W.J., Swanney, M.P., Donald, R.A.,Prickett, T.C. & Hellemans, J. (1998c) The effect of glycerol and desmopressinon exercise performance and hydrationin triathletes. Medicine and Science inSports and Exercise 30, 1263–1269.

Jacks, D.E., Sowash, J., Anning, J.,McGloughlin, T. & Andres, F. (2002)Effect of exercise at three exerciseintensities on salivary cortisol. Journal ofStrength and Conditioning Research 16,286–289.

Jard, S. (1988) Mechanisms of action of vasopressin and vasopressinantagonists. Kidney International.Supplement 26, S38–S42.

Jurimae, J. & Jurimae, T. (2001) Responsesof blood hormones to the maximalrowing ergometer test in college rowers.Journal of Sports Medicine and PhysicalFitness 41, 73–77.

Jurimae, J., Jurimae, T. & Purge, P. (2001)Plasma testosterone and cortisolresponses to prolonged sculling in malecompetitive rowers. Journal of SportsScience 19, 893–898.

Kaciuba-Uscilko, H., Porta, S., Nazar, K. et al. (1994) Effect of mild psychologicalstress on physiological responses toexercise in men. Journal of Physiology andPharmacology 45, 429–439.

multiset versus single-set heavy-resistance exercise protocols. CanadianJournal of Applied Physiology 22, 244–255.

Grossman, A. & Tsagarakis, S. (1989) The hunt for the CIA: factors whichdemonstrate corticotrophin-inhibitoryactivity. Journal of Endocrinology 123,169–172.

Grossman, A., Bouloux, P., Price, P. et al.(1984) The role of opioid peptides in thehormonal responses to acute exercise inman. Clinical Science 67, 483–491.

Häkkinen, K. & Pakarinen, A. (1995) Acutehormonal responses to heavy resistanceexercise in men and women at differentages. International Journal of SportsMedicine 16, 507–513.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi P.V. (1988a)Neuromuscular and hormonaladaptations in athletes to strengthtraining in 2 years. Journal of AppliedPhysiology 65, 2406–2412.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi, P.V. (1988b)Neuromuscular and hormonalresponses in elite athletes to twosuccessive strength training sessions in 1 day. European Journal of AppliedPhysiology 57, 133–139.

Harbach, H., Hell, K., Gramsch, C. et al.(2000) β-endorphin1–31 in the plasma ofmale volunteers undergoing physicalexercise. Psychoneuroendocrinology 25,551–562.

Harte, J.L., Eifert, G.H. & Smith, R. (1995)The effects of running and meditation on β-endorphin, corticotropin-releasinghormone and cortisol in plasma, and on mood. Biological Psychology 40,251–265.

Heitkamp, H.C., Huber, W. & Scheib, K. (1996) Beta-endorphin andadrenocorticotrophin after incrementalexercise and marathon runningafemaleresponses. European Journal of AppliedPhysiology 72, 417–424.

Hickson, R.C., Hidaka, K., Foster C.,Falduto, M.T. & Chatterton, R.T., Jr.(1994) Successive time courses ofstrength development and steroidhormone responses to heavy-resistancetraining. Journal of Applied Physiology 76,663–670.

Hoffman, J.R., Maresh, C.M., Armstrong,L.E. et al. (1994) Effects of hydration stateon plasma testosterone, cortisol andcatecholamine concentrations beforeand during mild exercise at elevatedtemperature. European Journal of AppliedPhysiology and Occupational Physiology69, 294–300.

Page 245: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 229

Kanaley, J.A., Weltman, J.Y., Pieper, K.S.,Weltman, A. & Hartman, M.L. (2001)Cortisol and growth hormone responsesto exercise at different times of day.Journal of Clinical Endocrinology andMetabolism 86, 2881–2889.

Keller-Wood, M.E. & Dallman, M.F. (1984)Corticosteroid inhibition of ACTHsecretion. Endocrine Reviews 5, 1–24.

Kenefick, R.W., Maresh, C.M., Armstrong,L.E. et al. (1998) Plasma testosterone andcortisol responses to training-intensityexercise in mild and hot environments.International Journal of Sports Medicine 19,177–181.

Kindermann, W., Schnabel, A., Schmitt,W.M. et al. (1982) Catecholamines,growth hormone, cortisol, insulin, andsex hormones in anaerobic and aerobicexercise. European Journal of AppliedPhysiology and Occupational Physiology49, 389–399.

Kirwan, J.P., Costill, D.L., Flynn, M.G. et al. (1988) Physiological responses tosuccessive days of intense training incompetitive swimmers. Medicine andScience in Sports and Exercise 20, 255–259.

Kjær, A. (1993) Vasopressin as aneuroendocrine regulator of anteriorpituitary hormone secretion. ActaEndocrinologica 129, 489–496.

Kjær, M., Bangsbo, J., Lortie, G. & Galbo,H. (1988) Hormonal response to exercisein humans: influence of hypoxia andphysical training. American Journal ofPhysiology 254, R197–R203.

Kraemer, R.R., Blair, S., Kraemer, G.R. & Castracane, V.D. (1989) Effects oftreadmill running on plasma β-endorphin, corticotropin, and cortisollevels in male and female 10 K runners.European Journal of Applied Physiology andOccupational Physiology 58, 845–851.

Kraemer, W.J., Fleck, S.J., Callister, R. et al.(1989) Training responses of plasma β-endorphin, adrenocorticotropin, andcortisol. Medicine and Science in Sportsand Exercise 21, 146–153.

Krieger, D.T., Allen, W., Rizzo, F. &Krieger, H.P. (1971) Characterization ofthe normal temporal pattern of plasmacorticosteroid levels. Journal of ClinicalEndocrinology and Metabolism 32,266–284.

Laatikainen, T.J. (1991) Corticotropin-releasing hormone and opioid peptidesin reproduction and stress. Annals ofMedicine 23, 489–496.

Langfort, J., Pilis, W., Zarzeczny, R., Nazar,K. & Kaciuba-Uscilko, H. (1996) Effect of low-carbohydrate-ketogenic diet onmetabolic and hormonal responses to

4000 m altitude. Hormone Research 41,225–229.

Martignoni, E., Appenzeller, O., Nappi, R.E. et al. (1997) The effects ofphysical exercise at high altitude onadrenocortical function in humans.Functional Neurology 12, 339–344.

el-Migdadi, F., Bashir, N., Hasan, Z., al-Hader, A.A. & Gharaibeh, M. (1996)Exercise at low altitude (Jordan Valley)causes changes in serum levels ofACTH, insulin, cortisol and lactate.Endocrine Research 22, 763–767.

Morley, J.E. (1981) The endocrinology of the opiates and opioid peptides.Metabolism 30, 195–209.

Mujika, I., Chatard, J.C., Padilla, S.,Guezennec, C.Y. & Geyssant, A. (1996)Hormonal responses to training and itstapering off in competitive swimmers:relationships with performance.European Journal of Applied Physiology 74,361–366.

Murray, R., Paul, G.L., Seifert, J.G. & Eddy,D.E. (1991) Responses to varying rates ofcarbohydrate ingestion during exercise.Medicine and Science in Sports and Exercise23, 713–718.

Nagel, D., Seiler, D. & Franz, H. (1992)Biochemical, hematological andendocrinological parameters duringrepeated intense short-term running in comparison to ultra-long-distancerunning. International Journal of SportsMedicine 13, 337–343.

Niess, A.M., Fehrenbach, E., Strobel, G. et al. (2003) Evaluation of stressresponses to interval training at low andmoderate altitudes. Medicine and Sciencein Sports and Exercise 35, 263–269.

O’Connor, P.J., Morgan, W.P. & Raglin, J.S.(1991) Psychobiologic effects of 3 d ofincreased training in female and maleswimmers. Medicine and Science in Sportsand Exercise 23, 1055–1061.

Op’t Eijnde, B. & Hespel, P. (2001) Short-term creatine supplementation does notalter the hormonal response to resistancetraining. Medicine and Science in Sportsand Exercise 33, 449–453.

Orth, D.N. (1992) Corticotropin-releasinghormone in humans. Endocrine Reviews13, 164–191.

Pelletier, G., Desy, L., Cote, J. & Vaudry, H.(1983) Immunocytochemical localizationof corticotropin-releasing factor-likeimmunoreactivity in the humanhypothalamus. Neuroscience Letters 41,259–263.

Pestell, R.G., Hurley, D.M. & Vandongen,R. (1989) Biochemical and hormonalchanges during a 1000 km

graded exercise in men. Journal ofPhysiology and Pharmacology 47, 361–371.

Lehmann, M., Gastmann, U., Petersen,K.G. et al. (1992) Training–overtraining:performance, and hormone levels, aftera defined increase in training volumeversus intensity in experienced middle-and long-distance runners. BritishJournal of Sports Medicine 26, 233–242.

Lehmann, M., Knizia, K., Gastmann, U. et al. (1993) Influence of 6-week, 6 daysper week, training on pituitary functionin recreational athletes. British Journal ofSports Medicine 27, 186–192.

Lehmann, M., Lormes, W., Opitzgress, A.et al. (1997) Training and overtraininga

an overview and experimental results in endurance sports. Journal of SportsMedicine and Physical Fitness 37, 7–17.

Lehmann, M., Foster, C., Dickhuth, H.H. & Gastmann, U. (1998) Autonomicimbalance hypothesis and overtrainingsyndrome. Medicine and Science in Sportsand Exercise 30, 1140–1145.

Liu, J.P., Robinson, P.J., Funder, J.W. & Engler, D. (1990) The biosynthesis and secretion of adrenocorticotropin by the ovine anterior pituitary ispredominantly regulated by argininevasopressin (AVP). Evidence thatprotein kinase C mediates the action ofAVP. Journal of Biological Chemistry 265,14 136–14 142.

Loucks, A.B. & Horvath, S.M. (1984)Exercise-induced stress responses ofamenorrheic and eumenorrheic runners.Journal of Clinical Endocrinology andMetabolism 59, 1109–1120.

Loucks, A.B., Mortola, J.F., Girton, L. &Yen, S.S. (1989) Alterations in thehypothalamic–pituitary–ovarian and the hypothalamic–pituitary–adrenalaxes in athletic women. Journal of ClinicalEndocrinology and Metabolism 68,402–411.

Luger, A., Deuster, P.A., Kyle, S.B. et al.(1987) Acute hypothalamic–pituitary–adrenal responses to the stress oftreadmill exercise. Physiologicadaptations to physical training. NewEngland Journal of Medicine 316,1309–1315.

Lutoslawska, G., Obminski, Z., Krogulski,A. & Sendecki, W. (1991) Plasma cortisoland testosterone following 19-km and42-km kayak races. Journal of SportsMedicine and Physical Fitness 31, 538–542.

Marinelli, M., Roi, G.S., Giacometti, M.,Bonini, P. & Banfi, G. (1994) Cortisol,testosterone, and free testosterone inathletes performing a marathon at

Page 246: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

230 chapter 17

ultramarathon. Clinical and ExperimentalPharmacology and Physiology 16, 353–361.

Petraglia, F., Barletta, C., Facchinetti, F. et al. (1988) Response of circulatingadrenocorticotropin, β-endorphin, β-lipotropin and cortisol to athleticcompetition. Acta Endocrinologica 118,332–336.

Petrides, J.S., Mueller, G.P., Kalogeras, K.T.et al. (1994) Exercise-induced activationof the hypothalamic–pituitary–adrenalaxis: marked differences in thesensitivity to glucocorticoidsuppression. Journal of ClinicalEndocrinology and Metabolism 79,377–383.

Petrides, J.S., Gold, P.W., Mueller, G.P. et al. (1997) Marked differences infunctioning of the hypothalamic–pituitary–adrenal axis between groupsof men. Journal of Applied Physiology 82,1979–1988.

Raastad, T., Bjoro, T. & Hallen, J. (2000)Hormonal responses to high- andmoderate-intensity strength exercise.European Journal of Applied Physiology 82,121–128.

Rahkila, P., Hakala, E., Salminen, K. &Laatikainen, T. (1987) Response ofplasma endorphins to running exercisesin male and female endurance athletes.Medicine and Science in Sports and Exercise19, 451–455.

Rich, P.A., Villani, R., Fulton, A. et al.(1992) Serum cortisol concentration and testosterone to cortisol ratio in eliteprepubescent male gymnasts duringtraining. European Journal of AppliedPhysiology and Occupational Physiology65, 399–402.

Rivier, C. & Vale, W. (1983) Modulation of stress-induced ACTH release bycorticotropin-releasing factor,catecholamines and vasopressin. Nature 305, 325–327.

Robertson, G.L. & Athar, S. (1976) Theinteraction of blood osmolality andblood volume in regulating plasmavasopressin in man. Journal of ClinicalEndocrinology and Metabolism 42,613–620.

Rolandi, E., Reggiani, E., Franceschini, R. et al. (1985) Comparison of pituitaryresponses to physical exercise in athletesand sedentary subjects. HormoneResearch 21, 209–213.

Ronkainen, H.R., Parkarinen, A.J. &Kauppila, A.J. (1986) Adrenocorticalfunction of female endurance runnersand joggers. Medicine and Science inSports and Exercise 18, 385–389.

and mental function in sedentary andendurance-trained elderly males.International Journal of Sports Medicine 20,159–166.

Suay, F., Salvador, A., Gonzalez-Bono, E. et al. (1999) Effects of competition and itsoutcome on serum testosterone, cortisoland prolactin. Psychoneuroendocrinology24, 551–566.

Sugimoto, T., Saito, M., Mochizuki, S. et al.(1994) Molecular cloning and functionalexpression of a cDNA encoding thehuman V1b vasopressin receptor.Journal of Biological Chemistry 269, 27 088–27 092.

Tabata, I., Atomi, Y., Mutoh, Y. &Miyashita, M. (1990) Effect of physicaltraining on the responses of serumadrenocorticotropic hormone duringprolonged exhausting exercise. EuropeanJournal of Applied Physiology andOccupational Physiology 61, 188–192.

Tabata, I., Ogita, F., Miyachi, M. &Shibayama, H. (1991) Effect of low bloodglucose on plasma CRF, ACTH, andcortisol during prolonged physicalexercise. Journal of Applied Physiology 71,1807–1812.

Tharp, G.D. & Buuck, R.J. (1974) Adrenaladaptation to chronic exercise. Journal ofApplied Physiology 37, 720–722.

Thoren, P., Floras, J., Hoffman, P. & Seals,D. (1990) Endorphins and exercise:physiological mechanisms and clinicalimplications. Medicine and Science inSports and Exercise 22, 417–428.

Thuma, J.R., Gilders, R., Verdun, M. &Loucks, A.B. (1995) Circadian rhythm of cortisol confounds cortisol responsesto exercise: implications for futureresearch. Journal of Applied Physiology 78,1657–1664.

Utter, A.C., Kang, J., Nieman, D.C. et al.(1999) Effect of carbohydrate ingestionand hormonal responses on ratings ofperceived exertion during prolongedcycling and running. European Journal ofApplied Physiology and OccupationalPhysiology 80, 92–99.

Uusitalo, A.L., Huttunen, P., Hanin, Y.,Uusitalo, A.J. & Rusko, H.K. (1998)Hormonal responses to endurancetraining and overtraining in femaleathletes. Clinical Journal of Sport Medicine8, 178–186.

Vale, W., Spiess, J., Rivier, C. & Rivier, J.(1981) Characterization of a 41-residueovine hypothalamic peptide thatstimulates secretion of corticotropin and β-endorphin. Science 78, 1394–1397.

Vanhelder, W.P., Radomski, M.W., Goode,

Ronsen, O., Haug, E., Pedersen, B.K. & Bahr, R. (2001a) Increasedneuroendocrine response to a repeatedbout of endurance exercise. Medicine andScience in Sports and Exercise 33, 568–575.

Ronsen, O., Holm, K., Staff, H. et al. (2001b)No effect of seasonal variation intraining load on immuno-endocrineresponses to acute exhaustive exercise.Scandinavian Journal of Medicine andScience in Sports 11, 141–148.

Schwarz, L. & Kindermann, W. (1989)Beta-endorphin, catecholamines, andcortisol during exhaustive enduranceexercise. International Journal of SportsMedicine 10, 324–328.

Schwarz, L. & Kindermann, W. (1990)Beta-endorphin, adrenocorticotropichormone, cortisol and catecholaminesduring aerobic and anaerobic exercise.European Journal of Applied Physiology andOccupational Physiology 61, 165–171.

Sgherza, A.L., Axen, K., Fain, R. et al. (2002)Effect of naloxone on perceived exertionand exercise capacity during maximalcycle ergometry. Journal of AppliedPhysiology 93, 2023–2028.

Silverman, H.G. & Mazzeo, R.S. (1996)Hormonal responses to maximal andsubmaximal exercise in trained anduntrained men of various ages. Journalsof Gerontology. Series A, Biological Sciences and Medical Sciences 51, B30–B37.

Singh, A., Petrides, J.S., Gold, P.W.,Chrousos, G.P. & Deuster, P.A. (1999)Differential hypothalamic–pituitary–adrenal axis reactivity to psychologicaland physical stress. Journal of ClinicalEndocrinology and Metabolism 84,1944–1948.

Smoak, B., Deuster, P., Rabin, D. &Chrousos, G. (1991) Corticotropin-releasing hormone is not the sole factor mediating exercise-inducedadrenocorticotropin release in humans.Journal of Clinical Endocrinology andMetabolism 73, 302–306.

Snegovskaya, V. & Viru, A. (1993) Steroidand pituitary hormone responses torowing: relative significance of exerciseintensity and duration and performancelevel. European Journal of AppliedPhysiology and Occupational Physiology67, 59–65.

Stewart, P.M. (2003) The adrenal cortex. In: Williams Textbook of Endocrinology(Larsen, P., Kronenberg, H., Melmed, S.& Polonsky, K., eds.). Saunders,Philadelphia: 491–551.

Struder, H.K., Hollmann, W., Platen, P. et al. (1999) Neuroendocrine system

Page 247: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and the hpa axis 231

R.C. & Casey, K. (1985) Hormonal andmetabolic response to three types ofexercise of equal duration and externalwork output. European Journal of AppliedPhysiology and Occupational Physiology54, 337–342.

Vasankari, T.J., Kujala, U.M., Heinonen,O.J. & Huhtaniemi, I.T. (1993) Effects of endurance training on hormonalresponses to prolonged physical exercisein males. Acta Endocrinologica 129,109–113.

Verde, T., Thomas, S. & Shephard, R.J.(1992) Potential markers of heavytraining in highly trained distancerunners. British Journal of Sports Medicine26, 167–175.

Wittert, G.A., Stewart, D.E., Graves, M.P. et al. (1991) Plasma corticotrophinreleasing factor and vasopressinresponses to exercise in normal man.Clinical Endocrinology 35, 311–317.

Wittert, G.A., Livesey, J.H., Espiner, E.A. & Donald. R.A. (1996) Adaptation of the hypothalamopituitary adrenal axisto chronic exercise stress in humans.Medicine and Science in Sports and Exercise28, 1015–1019.

Yanovski, J.A., Yanovski, S.Z., Boyle, A.J. et al. (2000) Hypothalamic–pituitary–adrenal axis activity during exercise in African American and Caucasianwomen. Journal of Clinical Endocrinologyand Metabolism 85, 2660–2663.

de Vries, W.R., Bernards, N.T., de Rooij,M.H. & Koppeschaar, H.P. (2000)Dynamic exercise discloses differenttime-related responses in stresshormones. Psychosomatic Medicine 62,866–872.

Wade, C.E. & Claybaugh, J.R. (1980)Plasma renin activity, vasopressinconcentration, and urinary excretoryresponses to exercise in men. Journal ofApplied Physiology 49, 930–936.

Whitnall, M.H., Smyth, D. & Gainer, H.(1987) Vasopressin coexists in half of thecorticotropin-releasing factor axonspresent in the external zone of themedian eminence in normal rats.Neuroendocrinology 45, 420–424.

Page 248: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

232

Introduction

Over the past 30 years three types of scientific studies have been conducted to investigate the highprevalence of menstrual disorders in athletes. Sur-veys have found the highest prevalences in compet-itive endurance, aesthetic and weight-class sportsand have led to several hypotheses about the causeof these disorders. Subsequent observational stud-ies comparing hormone measurements in amenor-rheic and eumenorrheic athletes contradicted mostof those hypotheses. Then prospective experimentscontrolling the administration of the two remain-ing hypothetical causal factors, exercise stress andenergy availability, have shown that low energyavailability (defined as dietary energy intake minusexercise energy expenditure) disrupts reproduct-ive function in physically active women and thatexercise has no suppressive effect on reproductivefunction beyond the impact of its energy cost onenergy availability.

In this research, most observational studies ofmenstrual disorders in athletes have focused onparticipants in aerobic sports, and most prospectiveexperiments have employed aerobic exercise train-ing to investigate the mechanism of these disorders.Therefore, very little information is available con-cerning whatever differences there may be betweenthe influences of aerobic and anaerobic exercise onthe prevalence and mechanism of menstrual dis-orders. However, since the prevalence of menstrualdisorders in female bodybuilders is high, and sincedietary restriction is common practice in bodybuild-ing, low energy availability probably accounts for

most cases of menstrual disorders in anaerobicsports, too.

Menstrual disorders caused by factors other than athletic training, including pregnancy, lacta-tion, eating disorders, pituitary tumors, hyperan-drogenism, polycystic ovary syndrome, depressionand various organic diseases, are beyond the scopeof this review, even though some (such as polycysticovary syndrome and anorexia nervosa) may be disproportionately represented amongst athletesdue to the self-selection of affected women intoactivities in which hyperandrogenism or low bodyweight, respectively, offer a competitive advantage.The subject of this review is the disruption of repro-ductive function by athletic training itself, the mech-anism of which has been traced to the failure offemale athletes to sufficiently increase their dietaryintake in compensation for their exercise energyexpenditure, and which may be prevented by dietaryreform without any moderation of their exerciseregimen.

Regulation of the reproductive system

The length and regularity of menstrual cycles vary considerably across the general population ofwomen as well as during an individual woman’sreproductive years (Treloar et al. 1967). The medianlength of the menstrual cycle amongst NorthAmerican adolescents is 33 days, and the medianstandard deviation of variations in the length frommonth to month is 7 days. By the age of 20 years, the median length and standard deviation havedecreased to 29 and 4 days, respectively. These

Chapter 18

Influence of Energy Availability on LuteinizingHormone Pulsatility and Menstrual Cyclicity

ANNE B. LOUCKS

Page 249: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 233

numbers continue to decline slowly until the age of 40 years when both numbers begin to increasegreatly during the years preceding menopause,when menstruation ceases permanently.

Moreover, the average age of menarche (i.e. thefirst menstrual cycle) has declined dramaticallyover the past 150 years in all Western societiesa

from an average of 17 years to 12.4 years (Styne &Grumbach 1991; Anderson, S.E. et al. 2003). Whatreactivates the reproductive system at puberty isunknown, just as what deactivates it in infancy andagain at menopause are also unknown.

The regulation of the menstrual cycle by thehypothalamic–pituitary–ovarian (HPO) axis includesboth negative and positive feedback mechanisms, aswell as inputs from the central nervous system andother systems at various levels within the axis. Theglands of the HPO axis secrete their hormonesrhythmically. Indeed, the secretion of gonadotropin-releasing hormone (GnRH) pulses into the pituitaryportal blood by neurons within the hypothalamusof the brain must occur at an optimal frequency ifthe axis as a whole is to function normally. TheseGnRH pulses stimulate the pulsatile secretion ofluteinizing hormone (LH) from the pituitary glandinto the blood. By sampling blood at 10 min inter-vals for 24 h, the frequency and amplitude of LHpulses can be studied.

In response to the proper pulsatile and monthlyrhythmic stimulation by LH and pituitary follicle-stimulating hormone (FSH), clusters of ovarian cells(‘ovarian follicles’) grow and secrete increasingamounts of estrogen. Gradually, too, one of the fol-licles becomes dominant and eventually the risingconcentration of estrogen in the blood exerts a positive feedback on LH secretion. In response, thepituitary gland secretes a surge of LH into the blood,causing the dominant follicle to rupture, therebyreleasing an egg cell for fertilization. The remainingcells of the dominant follicle then undergo rapidchemical and morphological changes to form thecorpus luteum, a body that begins secreting bothprogesterone and estrogen into the blood. The inter-val during which a dominant follicle develops, frommenses until ovulation, is known as the follicularphase of the menstrual cycle. The interval duringwhich the corpus luteum is active from ovulation

until the next menses is known as the luteal phase ofthe cycle.

Estrogen and progesterone have profound influ-ences on the uterine endometrium as well as manyother tissues in the body. Estrogen stimulates endo-metrial proliferation and progesterone causes it tobecome highly vascularized. These are necessary,hospitable conditions for the successful implanta-tion of a fertilized egg. If no fertilization occurs after several days, the ability of the corpus luteum tosecrete progesterone becomes exhausted, the struc-tural integrity of the endometrium collapses, andmenstruation ensues. Under normal circumstances,the secretory capacity of the corpus luteum is sus-tained long enough for the rapidly dividing cellsderived from a fertilized egg to become implantedin the endometrium 6 or 7 days after fertilization. If the secretory capacity of the corpus luteum isexhausted too soon, the endometrium sloughs offbefore implantation can occur. The likelihood of this increases when the luteal phase is shorter than 10 days. Thus, infertility can result from either thefailure of the ovary to release an egg for fertilizationor the failure of a fertilized egg to become properlyimplanted into the endometrium.

Characterization of female athletes

The ovarian axis

Amenorrheic athletes produce low levels of estrogenand progesterone every day indicating a completeabsence of follicular development, ovulation, andluteal function (Fig. 18.1, AA) (Loucks et al. 1989). Bycontrast, even the most eumenorrheic competit-ive athletes display extended follicular phases andabbreviated luteal phases with blunted progesteroneconcentrations (Fig. 18.1, CA) compared to eumen-orrheic sedentary women (Fig. 18.1, CS). Similarobservations have been made in eumenorrheicwomen running recreationally as little as 12 miles(20 km) per week (Ellison & Lager 1986; Broockset al. 1990; Pirke et al. 1990; De Souza et al. 1998).

The proximal cause of ovarian dysfunction inamenorrheic and eumenorrheic athletes is a disrup-tion of the pulsatile rhythm of LH concentrations inthe blood, upon which ovarian function critically

Page 250: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

234 chapter 18

depends (Veldhuis et al. 1985; Yahiro et al. 1987;Loucks et al. 1989; Laughlin & Yen 1996). In aeumenorrheic sedentary young woman, the 24-hLH profile in the early follicular phase is charac-terized by regular, high frequency pulses of lowamplitude (Fig. 18.2, CS) (Loucks et al. 1989). Duringsleep, the frequency slows and the amplitudeincreases. Eumenorrheic athletes display a slower,but still regular rhythm of larger pulses (Fig. 18.2,CA). Amenorrheic athletes display even fewerpulses, at irregular intervals (Fig. 18.2, AA).

Experimental administration of GnRH has de-monstrated that the disruption of LH pulsatility inamenorrheic and eumenorrheic athletes is causedby a disruption of GnRH pulsatility and not by apituitary disorder (Veldhuis et al. 1985; Loucks et al.1989). Therefore, the neuroendocrine mechanismsby which the GnRH pulse generator can be dis-rupted are the focus of much research.

The prevalence of amenorrhea in endurance, aesthetic and weight-class sports can be as much as10 times higher than in the general population (Otiset al. 1997). The less severe disorders of ovarianfunction (follicular and luteal suppression andanovulation) may display no menstrual symptomsat all so that affected women are entirely unaware of their condition until they undergo an endocrineworkup. Amongst eumenorrheic athletes, the incid-

ence of follicular and luteal suppression and ano-vulation appears to be extremely high. Repeatedendocrine workups have found that 79% of eumen-orrheic female runners were luteally suppressed oranovulatory in at least 1 month out of 3 (De Souzaet al. 1998).

Other endocrine axes

Mildly elevated resting serum cortisol levels inamenorrheic and eumenorrheic athletes (Ding et al.1988; Loucks et al. 1989; De Souza et al. 1991, 1994;Laughlin & Yen 1996) encouraged the hypothesisthat their reproductive disorders might be due tothe stress of exercise. Since cortisol is a glucoregulat-ory hormone activated by low blood glucose levels,however, these elevated cortisol levels might also be explained as part of the physiological response tochronic energy deficiency.

Indeed, more extensive endocrine observationsfound that amenorrheic athletes also display lowlevels of plasma glucose (Laughlin & Yen 1996),insulin (Laughlin & Yen 1996), insulin-like growthfactor I (IGF-I) (Zanker & Swaine 1998a), insulin-likegrowth factor I/insulin-like growth factor bindingprotein-1 (IGF-I/IGFBP-1) (an index of IGF-I bio-availability) (Laughlin & Yen 1996), leptin (Laughlin& Yen 1997; Thong et al. 2000) and triiodothyronine

CS

CA

AA

45

30

15

0

E 1G

ng

·mg

CR

–1

6

4

2

0

PdG

µg

·mg

CR

–1

Day from significant increase in PdG

–16 16–8 80

Fig. 18.1 Urinary estrone-glucuronide (E1G), an estradiolmetabolite, and pregnanediol-glucuronide (PdG), a progesteronemetabolite, over an entire menstrualcycle in cyclic sedentary women (CS)and cyclic athletes (CA), and over an entire month in amenorrheicathletes (AA). The mass of eachmetabolite (ngE1 and µPdG) excretedin overnight urine samples wasnormalized to the mass (mg) ofcreatinine (CR) excreted in the samesamples. The black and open bars atthe bottom of the figure indicate thedays of menses in the CS and and CA women, respectively, at thebeginning and the end of the cycle of observation. (From Loucks et al.1989.)

Page 251: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 235

(T3) (Myerson et al. 1991; Loucks et al. 1992; Zanker & Swaine 1998a, 1998b), as well as low restingmetabolic rates (Myerson et al. 1991). They also dis-play elevated growth hormone (GH) (Laughlin &

Yen 1996) in addition to the mildly elevated cortisollevels (Loucks et al. 1989; De Souza et al. 1991;Laughlin & Yen 1996). All these abnormalities aresigns of chronic energy deficiency.

Compared to eumenorrheic sedentary women,luteally suppressed eumenorrheic athletes also dis-play low levels of insulin (Laughlin & Yen 1996),leptin (Laughlin & Yen 1997) and T3 (De Souza et al.2003), and elevated levels of GH (Laughlin & Yen1996) and cortisol (Loucks et al. 1989; Laughlin &Yen 1996), but the magnitudes of these signs ofenergy deficiency are less extreme than in amenor-rheic athletes.

Energy intake and expenditure

Because of large variances between individuals, we were unable to distinguish between the dietaryand exercise habits and histories of the amenorrheicand eumenorrheic athletes that we studied (Louckset al. 1989, 1992). At that time, it did not occur to us to estimate each individual’s energy availability(dietary energy intake minus exercise energy expend-iture). Both groups of athletes reported their similarbody weights to be stable, despite dietary energyintakes indistinguishable from those of sedentarywomen (Loucks et al. 1989). That is, their dietaryenergy intakes were much less than would beexpected for their level of physical activity, a fea-ture commonly observed amongst athletic women(Drinkwater et al. 1984; Marcus et al. 1985; Nelsonet al. 1986; Kaiserauer et al. 1989; Myerson et al. 1991;Laughlin & Yen 1996).

Recently, extensive observational data on theenergy and carbohydrate intakes of athletes inmany sports have been compiled (Burke et al. 2001).If these data are to be believed, one observation isparticularly noteworthy: with the notable exceptionof cross-country skiers, female athletes consume ~ 30% less energy and carbohydrates⎯normalizedfor body weight⎯than do male athletes.

The repeatedly reported combination of a stablebody mass and an unexpectedly low dietary energyintake in female athletes is very controversial. Manyinvestigators have been skeptical of the dietaryrecords of female athletes, because studies com-paring such data to estimations or measurements

CS

CA

AA

AA

20

10

0

LH (

IU·L

–1)

20

10

0

LH (

IU·L

–1)

20

10

0

LH (

IU·L

–1)

20

10

0

LH (

IU·L

–1)

*

*

*

*

*

0800 08001600

Clock hours

2400

*

* *

*

*

*

* *

* **

* *

**

** * * * * * * *

Fig. 18.2 The 24-h pulsatile rhythms of luteinizinghormone (LH), expressed as international units per liter(IU·L–1), for a cyclic (regularly menstruating) sedentarywoman (CS), a cyclic athlete (CA) and two amenorrheicathletes (AA). Among amenorrheic athletes, theextensively quiescent, irregular pulsatile rhythms differedmarkedly. Pulsatile rhythms were considerably lessvariable among cyclic sedentary women and athletes.(From Loucks et al. 1989.)

Page 252: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

236 chapter 18

of their energy expenditure have repeatedly foundapparently huge negative energy balances, someexceeding 4 MJ·day–1 in athletes with stable bodyweights (Mulligan & Butterfield 1990; Wilmore et al.1992; Edwards et al. 1993; Beidleman et al. 1995;Trappe et al. 1997; Hill & Davies 2002). Such largediscrepancies have been interpreted as indicatingnot that female athletes are undernourished butrather that they grossly underreport their actualdietary intake. In support of this allegation, invest-igators have cited certain other special subpopula-tions that have been found to underreport (Wilmoreet al. 1992; Edwards et al. 1993). Actually, underre-porting of dietary intake is common in all popula-tions (Mertz et al. 1991) and a meta-analysis ofstudies comparing dietary assessments to measure-ments of energy expenditure by doubly labeledwater found that women do not underreport morethan men (Trabulsi & Schoeller 2001).

Other investigators have questioned the methodsused to measure energy intake and expenditure,and, indeed, the study that found virtually identicalenergy intakes in female and male cross-countryskiers took extraordinary pains to achieve accuratemeasurements of energy intake (Sjodin et al. 1994).As a result of such concerns, quantitative criteriahave been developed to assess whether reportedenergy intakes in studies of various numbers of sub-jects over various lengths of time pass what mightbe called the laugh test (Goldberg et al. 1991).

On the other hand, a stable body weight is notnecessarily proof of energy sufficiency, becausebehavior modification and endocrine-mediatedalterations in resting metabolic rate can counteractthe potential influences of dietary energy excess ordeficiency on body mass (Leibel et al. 1995). Energyintake and energy expenditure are also verydifficult to measure reliably, even with doublylabeled water. Considering the lack of confidence instudies of energy balance in athletes, therefore, it issurprising that investigations of energy intake andexpenditure in female athletes have not includedbiochemical measurements, because underreport-ing does not account for biochemical evidence ofenergy deficiency. As described above, metabolicsubstrates and hormones measured in amenorrheicand eumenorrheic athletes tell a consistent story of

chronic energy and carbohydrate deficiency result-ing in the mobilization of fat stores, the slowing ofmetabolic rate and a decline in glucose utilization,with more extreme abnormalities in amenorrheicathletes and less extreme abnormalities in eumenor-rheic athletes (Myerson et al. 1991; Loucks et al. 1992;Jenkins, P.J. et al. 1993; Laughlin & Yen 1996, 1997;De Souza et al. 2003). So, while some might suggestthat lower energy and carbohydrate intakes wouldbe appropriate for women if their energy and carbo-hydrate expenditures were less than those of men,biochemical data demonstrate that female athletesare, indeed, chronically energy deficient.

Clinical concerns

The stability of body weight in athletes whoseenergy intake is estimated to be much less than their energy expenditure has been attributed to an increase in metabolic ‘efficiency’ (Westerterp &Saris 1991; Westerterp et al. 1992), but ‘efficiency’ isnot the appropriate concept to apply to patholo-gical adjustments to chronic energy deficiency. Theoxidation of scarce metabolic fuels in the muscularwork of locomotion makes these fuels unavailablefor immune function, growth, tissue turnover, re-productive development and function, and otherimportant physiological functions. For example,50% of peak bone mass is deposited during adoles-cence, but the impairment of this process has led tosome young amenorrheic athletes having the bonedensities of 60-year-old women. Recently, a casestudy has been published documenting the 20-yearhistory of clinical osteoporosis in an amenorrheicathlete (Zanker et al. 2004).

Bone is continuously remodeled at millions oflocal sites by coupled processes of bone resorptionby osteoclast cells followed by bone formation byosteoblast cells. Normally, an increase in osteoclastactivity stimulates an increase in osteoblast activityand bone resorption and formation are said to becoupled, but under pathological circumstances theybecome uncoupled. When the activity of osteoclastsexceeds that of osteoblasts, the resulting net boneloss is irreversible, because once they have left abone remodeling site osteoblasts do not return tofinish the job of refilling it.

Page 253: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 237

At menopause, bone density declines because the normal suppression of osteoclast activity byestrogen is released, thereby increasing the rate ofbone resorption while bone formation is unaffected(Marcus et al. 1996) or increased (Nielsen et al. 2004).In younger women by contrast, energy deficiencyreduces the rate of bone formation by suppress-ing bone growth factors that stimulate osteoblastactivity, and when this energy deficiency is severeenough to induce amenorrhea, the suppression ofosteoclast activity by estrogen is also released,thereby increasing the rate of bone resorption. Thus,most studies of anorexia nervosa patients havefound both a reduction in bone formation as well asan increase in bone resorption (Stefanis et al. 1998;Caillot-Augusseau et al. 2000; Hotta et al. 2000; Audiet al. 2002; Gordon et al. 2002), although a few havefound only reduced formation (Soyka et al. 1999) orincreased resorption (Lennkh et al. 1999). Refeedinganorexia nervosa patients has been found either toincrease bone formation alone (Stefanis et al. 1998;Hotta et al. 2000) or to increase bone formation whilealso reducing resorption (Caillot-Augusseau et al.2000; Heer et al. 2002; Soyka et al. 2002).

Fewer studies of bone turnover have been con-ducted in amenorrheic athletes. Unfortunately, theresults of these studies have been less consistentthan those of anorexia nervosa patients, with resultsshowing no difference in either formation or resorp-tion (Hetland et al. 1993; Stacey et al. 1998), reducedformation (Okano et al. 1995), and reduced forma-tion and resorption but with resorption outweigh-ing formation (Zanker & Swaine 1998a). Therefore,more studies of bone turnover in amenorrheic ath-letes need to be conducted before we can speakconfidently about whether the mechanism of boneloss in amenorrheic athletes differs from that inanorexia nervosa patients. Such studies will need tobe interpreted with care, however, since markers ofbone turnover are systemic in nature and may notreveal local changes in bone turnover in the lumbarvertebrae and other trabecular sites where bone losscommonly occurs in amenorrheic athletes, espe-cially if bone density is simultaneously increasingelsewhere, such as in the heel.

In young athletic women, bone mineral densitydeclines by as much as 20% in proportion to the

number of menses that they have missed (Drinkwateret al. 1990), resulting in an increased rate of stressfractures (Warren et al. 2003). Oral contraceptiveshave been recommended to restore bone loss inamenorrheic athletes (Otis et al. 1997; Anderson, S.J.et al. 2000), but this treatment has failed to increasebone density in young amenorrheic women bymore than a few percent (Gulekli et al. 1994; Keen & Drinkwater 1997; Zanker et al. 2004). Restoringmenses has been more successful for increasingbone density in some women, but recovered amen-orrheic athletes have not kept pace with the bonegrowth of their eumenorrheic peers (Warren et al.2003). In recovered anorexia nervosa patients whohad been clinically well for 14–23 years, bone dens-ity in the femur remained 14% lower than in con-trol subjects (Hartman et al. 2000). The inability of estrogen to restore lost bone in amenorrheic ath-letes and anorexia nervosa patients may indicate apotentially reversible suppression of bone forma-tion caused by a continuing deficiency of nutritionalgrowth factors such as IGF-I, T3 and insulin, or itmay indicate that the bone loss is irreversible due tothe uncoupling of bone resorption and formation.

IGF-I and IGF-I/IGFBP-3 (another index of IGF-Ibioavailability) have been found to predict spinaland femoral neck bone mass in gymnasts and runners(Maddalozzo & Snow 2000). When anorexia ner-vosa patients with osteopenia were treated with acombination of oral contraceptives and recombin-ant IGF-I their bone density did increase more thanin a similar group who were administered oral con-traceptives alone (Grinspoon et al. 2000, 2002). Thebenefit was only a few percent, however, and it may have been illusory because the latter groupinexplicably reduced their dietary intake by 40%during the study resulting in a 15% decline in their endogenous IGF-1 production. Therefore, thesuperiority of the combined treatment has not beenestablished. This treatment has not yet been testedin amenorrheic athletes.

Skeletal demineralization and osteoporotic frac-tures are the most alarming clinical consequence of athletic amenorrhea. Osteoporotic spinal frac-tures result in permanent disabilities and chronicpain. Osteoporosis in a 20-year-old athlete is a dis-aster. Osteopenia is a disaster waiting to happen.

Page 254: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

238 chapter 18

Therefore, early detection and intervention are crit-ical for minimizing permanent skeletal damage.Physicians should not defer treatment until afterosteopenia and osteoporosis are manifest. TheAmerican College of Sports Medicine has publisheda position stand on the female athlete triad as a syndrome requiring prompt intervention to preventchronic undernutrition from inducing reproductivedisorders and skeletal demineralization (Otis et al.1997). The American Academy of Pediatrics haspublished a similar warning (Anderson, S.J. et al.2000). Since skeletal demineralization must pro-ceed for many months before a reduction in bonedensity is measurable, repeated measures of bio-chemical markers of bone turnover, which respondimmediately to undernutrition and hypoestrogen-ism should be considered as early indicators ofskeletal demineralization.

Of course, long before reductions in bone densityare measurable, the first clinical consequence ofamenorrhea is infertility, since amenorrheic womenare not developing egg cells that can be fertilized.Eumenorrheic physically active women with shortluteal phases and low progesterone levels may alsobe at risk for infertility due to failures of implanta-tion. Paradoxically, irregularly cycling and oligo-menorrheic athletes may be at increased risk forunintended pregnancies if they do not use con-traceptives, because their day of ovulation is lesspredictable than that of eumenorrheic women.

Another consequence of the hypoestrogenism inamenorrheic athletes is impaired endothelium-dependent arterial vasodilation (Hoch et al. 2003a,2003b), which reduces perfusion of working muscleand increases the risk of developing cardiovasculardisease, and which is restored by estrogen replace-ment therapy and by the return of regular menstrualcycles (Hoch et al. 2003b). Impaired skeletal muscleoxidative metabolism has also been reported inamenorrheic athletes, suggesting that they may beat a physiological disadvantage in their ability toperform repeated exercise bouts compared to theireumenorrheic competitors (Harber et al. 1998). Inanother report of estrogen-deficiency symptoms,75% of amenorrheic athletes reported vaginal dry-ness compared to only 7% of eumenorrheic athletes(Hammar et al. 2000).

Proposed mechanisms of reproductivedisturbances

As mentioned above, many competing hypothesesabout the cause and mechanism of reproductive dis-orders in athletes have been offered over the years.The three most prominent hypotheses are discussedbelow.

Body composition

The body composition hypothesis held that theovarian axis is disrupted when the amount ofenergy stored in the body as fat declines below acritical level (Frisch & McArthur 1974). Outside theresearch community this has been the most widelypublicized explanation for menstrual disorders inathletes, but it has been the least widely believedexplanation within the research community. Des-pite early associations of menarche and amenorrheawith body composition (Frisch & McArthur 1974),later observations of athletes did not consistentlyverify this association (e.g. Laughlin & Yen 1996),nor did they find the appropriate temporal relation-ship between changes in body composition andmenstrual function (for reviews see Scott & Johnston1982; Loucks & Horvath 1985; Sinning & Little 1987;Bronson & Manning 1991). Eumenorrheic andamenorrheic athletes span a common range of bodycomposition (Loucks et al. 1984; Sanborn et al. 1987;Laughlin & Yen 1997) leaner than most eumenor-rheic sedentary women. Among women distancerunners, energy balance is a better predictor ofestradiol levels (r = 0.88) than are body mass index(BMI) (r = 0.42) or percent body fat (r = 0.48) (Zanker& Swaine 1998c). Furthermore, if the growth andsexual development of young animals are blockedby dietary restriction, normal LH pulsatility re-sumes only a few hours after ad libitum feeding ispermitted, long before any change in body mass orcomposition (Bronson 1986). Further evidence thatbody composition does not play a causal role in themechanism of menstrual disorders was reported inan experiment on severely obese women (bodyweight ~ 130 kg; BMI ~ 47) (Di Carlo et al. 1999).Surgical reduction of their stomach volume reducedthe amount of food that these patients could eat, re-

Page 255: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 239

sulting in rapid weight loss and amenorrhea whilethe patients were still overweight (body weight ~ 97 kg; BMI ~ 35). Thus, the body compositionhypothesis confused causation with correlation,which occurs in athletes because a lean body com-position and menstrual disorders are both conse-quences of low energy availability.

Nevertheless, interest in the body compositionhypothesis was rejuvenated several years ago withthe discovery of the hormone leptin. Synthesizedand secreted by adipose tissue, leptin was origin-ally thought to communicate information about fatstores (Maffei et al. 1995). Later reports of leptinvarying profoundly before any changes in adiposityin response to fasting (Kolaczynski et al. 1996a;Weigle et al. 1997), dietary restriction (Weigle et al.1997), refeeding after dietary restriction (Kolaczynskiet al. 1996a; Jenkins, A.B. et al. 1997) and overfeeding(Kolaczynski et al. 1996b) led to the hypothesis thatleptin signals information about dietary intake, particularly carbohydrate intake (Jenkins, A.B. et al.1997). Then leptin was found to be regulated by thetiny flux of glucose through the hexosamine biosyn-thesis pathway in muscle and adipose tissue (Wanget al. 1998; Rossetti 2000; Obici et al. 2002; Ravussin2002). Since then, we have shown that the diurnalrhythm of leptin depends actually on energy avail-ability or more specifically on carbohydrate avail-ability (Hilton & Loucks 2000).

Exercise stress

The stress hypothesis held that the stress of exerciseactivates the adrenal axis, which disrupts the GnRHpulse generator by various mechanisms. To bemeaningfully distinct from the energy availabilityhypothesis, the stress hypothesis further impliedthat the adrenal axis is activated by some aspect ofexercise other than its energy cost.

Certainly, there are central and peripheral mech-anisms by which the adrenal axis can disrupt theovarian axis. Considerable animal research hasshown that GnRH neurons are disturbed by activa-tion of the hypothalamic–pituitary–adrenal axis via pathways involving corticotropin-releasing hor-mone (CRH) and endogenous opioid and proopi-omelanocortin-derived peptides, or by increased

cortisol negative feedback (Rivier & Rivest 1991;Chrousos & Gold 1992). Furthermore, early experi-ments by Selye (1939) induced anestrus and ovarianatrophy in rats by abruptly forcing them to runstrenuously for prolonged periods. Others inducedanestrus in rats by forced swimming (Asahina et al.1959; Axelson 1987), by forced running (Chattertonet al. 1990), and by requiring animals to run fartherand farther for smaller and smaller food rewards(Manning & Bronson 1989, 1991). Elevated cortisollevels in such studies were interpreted as signs ofstress, and the induced reproductive disorders werewidely interpreted as evidence that ‘exercise stress’had a counter-regulatory influence on the femalereproductive system. These experiments inducedextreme activations of the adrenal axis, however,raising cortisol concentrations by several hundredpercent, in contrast to the mild 10–30% elevationsseen in amenorrheic athletes (Loucks et al. 1989;Laughlin & Yen 1996), hypothalamic amenorrheapatients (Berga et al. 1989) and anorexia nervosapatients (Gold et al. 1986). Whether such mild eleva-tions in cortisol influence the GnRH pulse generatorwas entirely speculative.

Only one experiment had successfully employedexercise to induce menstrual disorders in eumenor-rheic women. That experiment (Bullen et al. 1985)imposed a high volume of aerobic exercise abruptly,in imitation of Selye (1939). It caused a large propor-tion of menstrual disorders in the first month, andan even larger proportion in the second. The dis-orders were more prevalent in a subgroup fed a con-trolled weight-loss diet than in another subgroupfed for weight maintenance, but even the weightmaintenance subgroup may have been underfed,since body mass is an unreliable indicator of energybalance (Leibel et al. 1995).

The first cracks in the stress hypothesis appearedwhen glucose administration during exercise wasfound to prevent the cortisol response to exercise inboth rats (Slentz et al. 1990) and in men (Tabata et al.1991) in the laboratory. This finding was laterconfirmed in a field experiment when the cortisolresponse to a strenuous, prolonged hill walk wasprevented by eating larger meals (Ainslie et al.2003). Then LH pulsatility was disrupted in habitu-ally sedentary, eumenorrheic women through a

Page 256: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

240 chapter 18

combination of dietary restriction and exercise(Williams et al. 1995), but this did not resolve theambiguity about whether exercise stress or energyavailability disrupts the reproductive system inexercising women.

Since all previous animal and human investiga-tions of the influence of the ‘activity stress paradigm’on reproductive function had confounded the stressof exercise with the stress of forcing animals to exer-cise and/or with energy deficiency, there was, infact, no unconfounded experimental evidence thatthe stress of exercise, independent of its energy cost,disrupts reproductive function in voluntarily exer-cising women. Therefore, we conducted an experi-ment to determine the independent effects ofexercise stress and energy availability on LH pul-satility (Loucks et al. 1998). We defined, measuredand controlled energy availability operationally asdietary energy intake minus exercise energy expend-iture. Needing an operational definition of exercisestress, however, we were confronted by a funda-mental problem. Despite 60 years of research onresponses to exercise stress, we could find no object-ive definition of exercise stress itself. So, we definedexercise stress as everything associated with exercise,except its energy cost.

Figure 18.3 shows the experimental design inwhich we assigned habitually sedentary women ofnormal body composition to sedentary or exercisinggroups and then administered balanced (45 kcal[188 kJ]·kg fat free mass [FFM]–1·day–1) and restricted10 kcal [42 kJ]·kg FFM–1·day–1) energy availabilitytreatments to them in random order under con-trolled conditions in the laboratory. The energyavailability of the balanced sedentary group wasachieved by feeding them 45 kcal [188 kJ]·kgFFM–1·day–1 of energy in the form of a clinical dietaryproduct. In the other trial, their energy availabil-ity was reduced by dietary restriction alone. Theexercising group expended 30 kcal [125 kJ]·kgFFM–1·day–1 of energy in supervised exercise on atreadmill in the laboratory. Their 10 kcal [42 kJ]·kgFFM–1·day–1 restricted energy availability wasachieved by feeding them a dietary energy intake of 40 kcal [167 kJ]·kg FFM–1·day–1 similar to thesedentary women in their balanced treatment. Theirbalanced energy availability was achieved by

increasing their dietary energy intake in compensa-tion for their exercise energy expenditure. After 4 days of these treatments, we drew blood samplesfrom the subjects at 10-min intervals for 24 h toassess LH pulsatility.

The results in Fig. 18.4 show that exercise stresshad no suppressive effect on LH pulse frequency,whereas low energy availability suppressed LHpulse frequency, regardless of whether the lowenergy availability was caused by dietary energyrestriction alone or by exercise energy expenditurealone. We also obtained similar results (not shown)when half of the reduction in energy availabilitywas caused by dietary energy restriction and half byexercise energy expenditure. Low energy availabil-ity also suppressed T3, insulin, IGF-I and leptin(Hilton & Loucks 2000) while increasing GH andcortisol in a pattern very reminiscent of amenorrh-eic and luteally suppressed eumenorrheic athletes.

Unexpectedly, the effects of low energy availabil-ity on LH pulse frequency and on the metabolic hor-mones were smaller in the exercising women than inthe dietarily restricted women, even though theirbalanced and low energy availabilities were exactly

I E

S

A I

B

D

E

X

A

90

45

0

–45

90

45

0

–45

90

45

0

–45

90

45

0

–45

kcal

·kg

FFM

–1

Fig. 18.3 Experimental design. Dietary energy intake (I)and exercise energy expenditure (E) were controlled toachieve balanced (B = 45 kcal·kg FFM–1·day–1) anddeprived (D = 10 kcal·kg FFM–1·day–1) energy availability(A = I − E) treatments. Deprived energy availability wasachieved by dietary restriction alone in sedentary women(S) and by exercise energy expenditure alone in exercisingwomen (X) (1 kcal = 4.18 kJ). (From Loucks et al. 1998.)

Page 257: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 241

matched. This surprised us, because no-one hadever hypothesized that exercise would be protectiveof reproductive function. Further investigationrevealed that the exercising women had a highercarbohydrate availability (defined observationallyas dietary carbohydrate intake minus carbohydrateoxidation during exercise), due to a glucose-sparingalteration in skeletal muscle fuel selection duringenergy deprivation. As a result, the carbohydrateavailabilities of the exercising and sedentary womenwere above and below the brain’s daily glucoserequirement, respectively.

Since this experiment, amenorrhea has beeninduced in monkeys by training them to run volun-tarily on a motorized treadmill for longer and longerperiods while their food intake remained constant(Williams et al. 2001a). The monkeys became amen-orrheic abruptly in 7–24 months, after one or twocycles of luteal suppression. When the diet of half of the monkeys was then supplemented withoutany moderation of their exercise regimen, theirmenstrual cycles were restored (Williams et al.2001b). The rapidity of recovery was directly relatedto the number of calories consumed.

Further data undermining the stress hypothesishave been reported in a study of young male sol-diers participating in the 8-week US Army Rangertraining course (Friedl et al. 2000). This course is

divided into four 2-week phases in forest, desert,mountain and swamp environments during whichtrainees undergo daily military skill training, 8–12 km patrols carrying 32 kg rucksacks, sleep depri-vation (~ 3.6 h of sleep per night) and dietaryintakes during alternate weeks of ~ 8.4 and ~ 21 MJper day. During the course, trainees lost ~ 12 kg ofbody weight. Blood sampling at the end of eachweek revealed that T3, IGF-I and testosterone levelsfell ~ 20%, ~ 50% and ~ 70%, respectively, duringweeks on diets of 8.4 MJ·day–1 and returned to normal initial levels during alternate weeks on dietsof 21 MJ·day–1, despite continued exposure to allother training stresses. Thus, exercise appears tohave no deleterious effect on reproductive functionin either men or women apart from the impact of itsenergy cost on energy availability. If the adrenalaxis disrupts the GnRH pulse generator in athletes,it probably does so by mediating the influence ofenergy availability.

Energy availability

The energy availability hypothesis recognizes thatmammals partition energy amongst six major meta-bolic activities: cellular maintenance, thermoregula-tion, immunity, locomotion, growth and reproduction(Wade & Schneider 1992). The expenditure ofenergy in one function, such as locomotion, makes itunavailable for others, such as reproductive devel-opment and function. Specifically, this hypothesisholds that failure to provide sufficient metabolicfuels to meet the energy requirements of the braincauses an alteration in brain function that disruptsthe GnRH pulse generator. In this regard, it isimportant to remember that because fatty acids donot cross the blood–brain barrier, the brain relies onglucose for energy. Furthermore, the brain has noglucose storage capacity, and in humans the brain isso large and so metabolically active that its dailyenergy requirement exceeds the entire liver glyco-gen storage capacity (Bursztein et al. 1989). In addi-tion, because skeletal muscle lacks the enzyme toreturn glucose derived from muscle glycogen to thebloodstream, muscle glycogen stores are not avail-able to the brain. By contrast, skeletal muscle doeshave access to liver glycogen stores. Therefore,

Puls

es/2

4h

S X

20

15

10

5

0S

*

*

*

X

0

–1

–2

–3

–4

–5

–6

–7

Fig. 18.4 Left: luteinizing hormone (LH) pulse frequencyin sedentary (S) and exercising (X) women with the sameenergy availability. Right: reduction in LH pulsefrequency caused by low energy availability in sedentary(S) and exercising (X) women. * = p < 0.01. (From Loucks2004; adapted from Loucks et al. 1998.)

Page 258: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

242 chapter 18

working skeletal muscle competes directly againstthe brain for all available carbohydrate stores in thebody. In a marathon race, working muscle oxidizesas much glucose in 2 h as the brain needs in a week.

Considerable data from biological field trials sup-port the hypothesis that mammalian reproductivefunction depends on energy availability, particu-larly in women (for reviews see Bronson & Manning1989; Wade & Schneider 1992; Bronson & Heideman1994; Wade et al. 1996; Schneider & Wade 2000).Anestrus has been induced in Syrian hamsters byfood restriction, the administration of pharmacolo-gical blockers of carbohydrate and fat metabolism,insulin administration (which shunts metabolicfuels into storage) and cold exposure (which con-sumes metabolic fuels in thermogenesis) (Wade &Schneider 1992). Disruptive effects on the reproduct-ive system were independent of body size and composition.

Considerable laboratory research suggests thatGnRH neuron activity and LH pulsatility are regu-lated by brain glucose availability via two separatemechanisms involving the area postrema (AP) inthe caudal brain stem and the vagus nerve (Knobil1990; Minami et al. 1995; Levin et al. 1999; Muroyaet al. 1999; Wade & Jones 2003). Glucose-sensingneurons in the AP of the hindbrain appear to trans-mit information to the GnRH pulse generator in the arcuate nucleus of the hypothalamus in the forebrain via neurons containing catecholamines,neuropeptide Y and CRH. These glucose-sensingneurons are activated by fasting (Mizuno et al. 1999),due in part to reductions in the inhibitory influencesof insulin (Schwartz et al. 1992) and leptin (Mizunoet al. 1998) as well as glucose.

Having demonstrated that low energy avail-ability, not exercise stress, disrupts LH pulsatility in exercising women, we investigated the dose–response relationship between energy availabilityand LH pulsatility and between energy availabilityand metabolic substrates and hormones in exercis-ing women (Loucks & Thuma 2003). Energy avail-ability was set at 10, 20, 30 and 45 kcal [42, 84, 125and 188 kJ]·kg FFM–1·day–1 by having all subjectsperform 16 kcal [63 kJ]·kg FFM–1·day–1 of exercise at70% Vo2max while consuming 25, 35, 45 or 60 kcal[104, 146, 188, or 251 kJ]·kg FFM–1·day–1 of dietary

energy. All subjects were administered the balancedenergy availability treatment (45 kcal [188 kJ]·kgFFM–1·day–1) and one of the restricted energy avail-ability treatments in random order.

Figure 18.5 shows the dose-dependent effects of energy availability on LH pulsatility. LH pulsefrequency was suppressed and pulse amplitudewas increased below a threshold of energy avail-ability at ~ 30 kcal [125 kJ]·kg FFM–1·day–1, suggest-ing that athletes may be able to prevent menstrualdisorders by maintaining energy availabilities above30 kcal [125 kJ]·kg FFM–1·day–1.

We also found that the disruption of LH pulsat-ility is substantially more extreme in women withshort luteal phases (Fig. 18.6). If the latter finding is confirmed through further investigations, thescreening of women for luteal length may be a convenient way to identify those who need to takeextra care to avoid falling below the threshold ofenergy availability needed to maintain normal LHpulsatility.

Statistical analysis showed that the dose-dependent effects on LH pulsatility were similar to those on the metabolic substrates glucose and

Effe

cts

(%)

0

Amplitude/3

Frequency

10 20 30Energy availability (kcal·kg FFM–1·day–1)

40 50

50

25

0

–25

–50

Fig. 18.5 Dose-dependent effects of restricted energyavailability on luteinizing hormone (LH) pulse amplitude(� top) and frequency (� bottom). Effects are expressedrelative to values at 45 kcal·kg FFM–1·day–1. Effects on LHpulse amplitude are divided by three for graphicalsymmetry. As energy availability declines from 45 kcal·kgFFM–1·day–1, effects occur below 30 kcal·kg FFM–1·day–1

and become more extreme as energy availability is furtherreduced (1 kcal = 4.18 kJ). (From Loucks & Thuma 2003.)

Page 259: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 243

β-hydroxybutyrate and to the metabolic hormonesGH and cortisol, and unlike those of the othermetabolic hormones including insulin, IGF-I, T3and leptin. These results support the hypothesisthat reproductive function reflects the availability of metabolic fuels, especially glucose, which may besignaled in part by activation of the adrenal axis.The role of leptin in mediating the influence ofenergy metabolism on the reproductive axis remainscontroversial with both proponents (Hileman et al.2000) and skeptics (Schneider & Wade 2000). Thecentral melanocortin mechanism by which leptininhibits food intake does not appear to mediate lep-tin’s stimulatory effect on the GnRH pulse generator(Hohmann et al. 2000).

The maintenance of normal LH pulsatility in thisexperiment despite a 33% restriction of energyavailability to 30 kcal [125 kJ]·kg FFM–1·day–1

demonstrated for the first time that LH pulsatility isnot simply proportional to energy availability.Rather, the regulation of the reproductive system inwomen seems to be robust against reductions inenergy availability as large as 33%. Since the exer-

cise energy expenditure in this experiment was ~ 836 kcal [3.5 MJ]·day–1, these results suggest thatmany women may be able to maintain normal LHpulsatility while running up to 8 miles (13 km) a dayas long as they do not simultaneously reduce theirdietary energy intake below 45 kcal [188 kJ]·kgFFM–1·day–1. If they do reduce their dietary energyintake, as many exercising women do, then they riskfalling below the threshold of energy availabilityneeded to maintain normal LH pulsatility.

We also determined the dose-dependent effects of energy availability on biochemical markers ofbone turnover in this experiment (Ihle & Loucks2004). We found that bone resorption increased andbecame uncoupled from a suppression of bone for-mation only when energy availability was res-tricted severely enough to suppress estradiol (i.e. to10 kcal [42 kJ]·kg FFM–1·day–1). If left to continue,such uncoupling may cause irreversible reductionsin bone density. By contrast, bone formation wasimpaired by much less severe restrictions of energyavailability (i.e. as high as 30 kcal [125 kJ]·kgFFM–1·day–1), a level not at all unusual in weightcontrol programs, in close association with theeffects of low energy availability on insulin, IGF-Iand T3. Such reductions in the rate of bone forma-tion may prevent even regularly menstruating, phys-ically active women from achieving their geneticpotential for peak bone mass.

Recently, a group of physically active universitywomen displayed high scores on a test of dietaryrestraint without any of them reporting irregularmenstrual cycles (McLean et al. 2001). From the dataprovided, we estimate that their energy availabilit-ies were reduced only ~ 22% compared to similarwomen with low restraint scores. Similarly, monkeysmaintained on a 30% restricted diet for 6 yearsshowed no disruption of menstrual cycling orreproductive hormones and no reduction in bonemineral density, despite a fat mass 46% lower thanthat in monkeys fed a control diet (Lane et al. 2001).Since their body mass declined during the study,their dietary intake normalized to their body masshad, in fact, declined by only ~ 20%. Recent cross-sectional comparisons of estimated energy avail-ability in athletes also support the notion thatmenstrual function is disrupted at the threshold of

Effe

cts

(%)

0

A11/3

A > 11/3

F > 11

F11

10 20 30Energy availability (kcal·kg FFM–1·day–1)

40 50

75

50

25

0

–25

–50

–75

Fig. 18.6 Dose-dependent effects of restricted energyavailability on luteinizing hormone (LH) pulse amplitude(�, � top) and frequency (�,� bottom) in subgroups ofwomen with luteal phases of 11 days (�, �) and > 11 days(�, �). Women with shorter luteal phases (< 11 days) hadbeen excluded from participation in the experiment.Effects are relative to values at 45 kcal·kg FFM–1·day–1.Effects on LH pulse amplitude are divided by three forgraphical symmetry. Women with luteal phases of 11 daysdisplayed substantially more extreme disruption of LHpulsatility (1 kcal = 4.18 kJ). (From Loucks & Thuma 2003.)

Page 260: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

244 chapter 18

energy availability needed to maintain normal LHpulsatility (Thong et al. 2000). Amenorrheic athleteswere estimated to habitually self-administer anenergy availability of ~ 16 kcal [67 kJ]·kg FFM–1·day–1,while eumenorrheic athletes habitually self-admin-istered ~ 30 kcal [125 kJ]·kg FFM–1·day–1.

Implications for athletic training

Research done to date suggests that athletes may beable to prevent or reverse menstrual disorders byincreasing their dietary energy intake without anymodification of their exercise regimen. In short-term experiments, an energy availability of 30 kcal[125 kJ]·kg FFM–1·day–1 appears to be sufficient topreserve normal LH pulsatility. More short-termexperiments are needed to determine the independ-ent effects of carbohydrate and energy availabilityon LH pulsatility, to identify the exact physiologicalmechanism by which low energy or carbohydrateavailability disrupts LH pulsatility, and to investig-ate further whether low energy availability causesmore extreme disruptions of reproductive functionin certain identifiable women. More prolongedexperiments are needed to verify that short-termeffects on LH pulsatility are predictive of chroniceffects on ovarian function, and to identify practical,effective and acceptable interventions for prevent-ing and reversing menstrual disorders in athletes.Research is also needed to investigate how bodyweight is maintained in chronically energy-deficientathletes.

Part of the nutritional challenge for athletes is that‘there is no strong biological imperative to matchenergy intake to activity-induced energy expend-iture’ (Blundell & King 1999, p. 5581). Experimentalfood deprivation increases hunger, but the sameenergy deficit produced by exercise energy expend-iture does not (Hubert et al. 1998). Studies haveshown that hunger is suppressed briefly by a singlebout of intense exercise (Blundell & King 1998), andtwo bouts of intense exercise in a single day inducesno increase in ad libitum food intake on that or thefollowing 2 days (King et al. 1997). Furthermore,large shifts in carbohydrate and fat oxidation(Stubbs et al. 1995a, 1995b) and in glycogen stores(Snitker et al. 1997) produce no changes in ad libitum

macronutrient intake. Even a 20% increase in energyexpenditure during 40 weeks of marathon traininginduced no increase in energy intake (Westerterpet al. 1992). In our own laboratory, women say thatthey have to force themselves to eat far beyond their appetites to consume the amount of food thatcompensates their dietary energy intake for theirexercise energy expenditure and to prevent the disruption of LH pulsatility. Other investigatorshave had to offer exercising amenorrheic monkeysspecial treats to induce them to increase their energyintake enough to restore their menstrual cycles(Williams et al. 2001b). Consequently, to improvetheir performance while protecting their health, athletes must learn to eat by discipline instead ofappetite.

Complicating that discipline, another part of thenutritional challenge for athletes is that energy balance is not the objective of athletic training.Athletic performance is maximized, in part, by asport-specific (and in team sports, position-specific)optimum body size, body composition and mix of stored metabolic fuels. Therefore, much of an athlete’s training aims to modify her body toachieve these objectives. It is also important to knowthat macronutrients are metabolized differently and stored separately so that the conversion of onemacronutrient into another for storage does not rep-resent important metabolic pathways (Flatt 1988).Therefore, an athlete needs to manage fat, proteinand carbohydrate balances separately to optimizetheir pursuit of sport-specific body size, body com-position and energy store objectives.

As part of this discipline, athletes would benefitfrom monitoring biomarkers of their progress andpitfalls on the path toward their particular bodysize, body composition and energy store object-ives. Much applied research is needed to validatethe utility of such biomarkers. Ideally, a single measurement of a biomarker would provide thedesired information accurately, unambiguously,inexpensively, conveniently, non-invasively, safely,privately and quickly without intentional or un-intentional confounding by other factors. Few, ifany, biomarkers fulfill all these criteria. An obviouscandidate for monitoring progress in reducing fatmass is skinfold thickness, which is simple, direct,

Page 261: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 245

immediate and inexpensive, and perhaps goodenough. We are unaware of any inexpensive, non-invasive method for assessing muscle and liverglycogen stores, which are essential for both athleticperformance and reproductive health, but a con-venient method for assessing carbohydrate defici-ency is readily available. In our experience, urinaryketones identify carbohydrate-deficient individualsalmost as reliably at 30 kcal [125 kJ]·kg FFM–1·day–1

as they do at 10 kcal [42 kJ]·kg FFM–1·day–1. A singleprolonged exercise bout on an energy-restricted dietis sufficient to elevate plasma ketones (Ainslie et al.2003). Athletes can purchase ‘keto-sticks’ inexpens-ively in most pharmacies to monitor their urinaryketones in the privacy of their own homes.

The health of female athletes has always been a high priority of women’s sports, but undernutri-tion has become standard practice, especially inendurance, aesthetic and weight-class sports. Ifequally pervasive voluntary reforms by athletes,coaches and judges are unlikely, then mandatory

institutional reforms like Rule 3 in National Col-legiate Athletic Association (NCAA) men’s wrestling(NCAA 2002) may be necessary to protect the healthof female athletes. Rule 3 restricts weight loss object-ives and sets individualized minimum weights forcompetition. In so doing, it prevents widespreadweight loss practices that had previously placed thehealth of participants at risk.

Acknowledgments

This research was supported in part by the US ArmyMedical Research Acquisition Activity (USAM-RAA) grant #DAMD 17-95-1-5053, and in part byGrant M01 RR00034 from the General ClinicalResearch Branch, Division of Research Resources,NIH. The content of the information presented inthis manuscript does not necessarily reflect the position or the policy of the government, and noofficial endorsement should be inferred.

References

Ainslie, P.N., Campbell, I.T., Frayn, K.N. et al. (2003) Physiological, metabolic, and performance implications of aprolonged hill walk: influence of energyintake. Journal of Applied Physiology 94(3),1075–1083.

Anderson, S.E., Dallal, G.E. & Must, A.(2003) Relative weight and raceinfluence average age at menarche:Results from two nationallyrepresentative surveys of US girlsstudied 25 years apart. Pediatrics 111,844–850.

Anderson, S.J., Griesemer, B.A., Johnson,M.D. et al. (2000) American Academy of Pediatrics. Committee on SportsMedicine and Fitness. Medical concernsin the female athlete. Pediatrics 106(3),610–613.

Asahina, K., Kitahara, F., Yamanaka, M. &Akiba, T. (1959) Influence of excessiveexercise on the structure and function ofrat organs. Japanese Journal of Physiology9, 322–326.

Audi, L., Vargas, D.M., Gussinye, M. et al. (2002) Clinical and biochemicaldeterminants of bone metabolism andbone mass in adolescent female patientswith anorexia nervosa. Pediatric Research51(4), 497–504.

Axelson, J.F. (1987) Forced swimmingalters vaginal estrous cycles, bodycomposition, and steroid levels withoutdisrupting lordosis behavior or fertilityin rats. Physiology and Behavior 41(5),471–479.

Beidleman, B.A., Puhl, J.L. & De Souza,M.J. (1995) Energy balance in femaledistance runners. American Journal ofClinical Nutrition 61(2), 303–311.

Berga, S.L., Mortola, J.F., Girton, L. et al.(1989) Neuroendocrine aberrations inwomen with functional hypothalamicamenorrhea. The Journal of ClinicalEndocrinology and Metabolism 68,301–309.

Blundell, J.E. & King, N.A. (1998) Effects of exercise on appetite control: loosecoupling between energy expenditureand energy intake. International Journal of Obesity and Related Metabolic Disorders22 (suppl. 2), S22–S29.

Blundell, J.E. & King, N.A. (1999) Physicalactivity and regulation of food intake:current evidence. Medicine and Science inSports and Exercise 31(11), S573–S583.

Bronson, F.H. (1986) Food-restricted,prepubertal, female rats: rapid recoveryof luteinizing hormone pulsing withexcess food, and full recovery of

pubertal development withgonadotropin-releasing hormone.Endocrinology 118(6), 2483–2487.

Bronson, F.H. & Heideman, P.D. (1994)Seasonal regulation of reproduction in mammals. In: The Physiology ofReproduction, Vol. 2 (Knobil, E. & Neill,J., eds.). Raven Press, New York:541–583.

Bronson, F.H. & Manning, J.M. (1989) Foodconsumption, prolonged exercise, andLH secretion in the peripubertal femalerat. In: The Menstrual Cycle and ItsDisorders (Pirke, K.M., Wuttle, W. &Schweiger, U., eds.). Springer-Verlag,Berlin: 42–29.

Bronson, F.H. & Manning, J.M. (1991) The energetic regulation of ovulation: a realistic role for body fat. Biology ofReproduction 44(6), 945–950.

Broocks, A., Pirke, K.M., Schweiger, U. et al. (1990) Cyclic ovarian function inrecreational athletes. Journal of AppliedPhysiology 68(5), 2083–2086.

Bullen, B.A., Skrinar, G.S., Beitins, I.Z. et al. (1985) Induction of menstrualdisorders by strenuous exercise inuntrained women. New England Journalof Medicine 312(21), 1349–1353.

Burke, L.M., Cox, G.R., Culmmings, N.K.

Page 262: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

246 chapter 18

et al. (2001) Guidelines for dailycarbohydrate intake: do athletes achievethem? Sports Medicine 31(4), 267–299.

Bursztein, S., Elwyn, D.H., Askanazi, J. & Kinney, J.M. (1989) Energy Metabolism,Indirect Calorimetry, and Nutrition.Williams & Wilkins, Baltimore, MD: 146.

Caillot-Augusseau, A., Lafage-Proust,M.H., Margaillan, P.H. et al. (2000)Weight gain reverses bone turnover and restores circadian variation of boneresorption in anorexic patients. ClinicalEndocrinology 52(1), 113–121.

Chatterton, R.T., Jr., Hartman, A.L., Lynn,D.E. & Hickson, R.C. (1990) Exercise-induced ovarian dysfunction in the rat.Proceedings of the Society for ExperimentalBiology and Medicine 193(3), 220–224.

Chrousos, G. P. & Gold, P.W. (1992) Theconcepts of stress and stress systemdisorders. Journal of the American MedicalAssociation 267, 1244–1252.

De Souza, M.J., Maguire, M.S., Maresh,C.M. et al. (1991) Adrenal activation and the prolactin response to exercise ineumenorrheic and amenorrheic runners.Journal of Applied Physiology 70(6),2378–2387.

De Souza, M.J., Luciano, A.A., Arce, J.C.,Demers, L.M. & Loucks, A.B. (1994)Clinical tests explain blunted cortisolresponsiveness but not mildhypercortisolism in amenorrheicrunners. Journal of Applied Physiology76(3), 1302–1309.

De Souza, M.J., Miller, B.E., Loucks, A.B. et al. (1998) High frequency of lutealphase deficiency and anovulation inrecreational women runners: bluntedelevation in follicle-stimulatinghormone observed during luteal-follicular transition. Journal of ClinicalEndocrinology and Metabolism 83(12),4220–4232.

De Souza, M.J., Van Heest, J., Demers, L.M.& Lasley, B.L. (2003) Luteal phasedeficiency in recreational runners:evidence for a hypometabolic state.Journal of Clinical Endocrinology andMetabolism 88(1), 337–346.

Di Carlo, C., Palomba, S., De Fazio, M. et al. (1999) Hypogonadotropichypogonadotropism in obese womenafter biliopancreatic diversion. Fertilityand Sterility 72(5), 905–909.

Ding, J.-H., Scheckter, C.B., Drinkwater,B.L., Soules, M.R. & Bremner, W.J. (1988) High serum cortisol levels inexercise-associated amenorrhea. Annalsof Internal Medicine 108, 530–534.

Drinkwater, B.L., Nilson, K., Chesnut,C.H., 3rd et al. (1984) Bone mineral

contraceptive administration on bonedensity in anorexia nervosa. Journal ofClinical Endocrinology and Metabolism87(6), 2883–2891.

Gulekli, B., Davies, M.C. & Jacobs, H.S.(1994) Effect of treatment on establishedosteoporosis in young women withamenorrhoea. Clinical Endocrinology41(3), 275–281.

Hammar, M.L., Hammar-Henriksson,M.B., Frisk, J., Rickenlund, A. & Wyon,Y.A. (2000) Few oligo-amenorrheicathletes have vasomotor symptoms.Maturitas 34(3), 219–225.

Harber, V.J., Petersen, S.R. & Chilibeck,P.D. (1998) Thyroid hormoneconcentrations and muscle metabolismin amenorrheic and eumenorrheicathletes. Canadian Journal of AppliedPhysiology 23, 293–306.

Hartman, D., Crisp, A., Rooney, B. et al.(2000) Bone density of women who have recovered from anorexia nervosa.International Journal of Eating Disorders28(1), 107–112.

Heer, M., Mika, C., Grzella, I., Drummer,C. & Herpertz-Dahlmann, B. (2002)Changes in bone turnover in patientswith anorexia nervosa during 11 weeksof inpatient dietary treatment. ClinicalChemistry 48(5), 754–760.

Hetland, M.L., Haarbo, J., Christiansen, C.& Larsen, T. (1993) Running inducesmenstrual disturbances but bone mass is unaffected, except in amenorrheicwomen. American Journal of Medicine95(1), 53–60.

Hileman, S.M., Pierroz, D.D. & Flier, J.S.(2000) Leptin, nutrition, andreproduction: timing is everything.Journal of Clinical Endocrinology andMetabolism 85(2), 804–806.

Hill, R.J. & Davies, P.S. (2002) Energyintake and energy expenditure in elitelightweight female rowers. Medicine andScience in Sports and Exercise 34(11),1823–1829.

Hilton, L.K. & Loucks, A.B. (2000) Lowenergy availability, not exercise stress,suppresses the diurnal rhythm of leptinin healthy young women. AmericanJournal of Physiology. Endocrinology andMetabolism 278(1), E43–E49.

Hoch, A.Z., Dempsey, R.L., Carrera, G.F. et al. (2003a) Is there an associationbetween athletic amenorrhea andendothelial cell dysfunction? Medicineand Science in Sports and Exercise 35(3),377–383.

Hoch, A.Z., Jurva, J., Staton, M. et al.(2003b) Is endothelial dysfunction that is associated with athletic amenorrhea

content of amenorrheic andeumenorrheic athletes. New EnglandJournal of Medicine 311(5), 277–281.

Drinkwater, B.L., Bruemner, B. & Chesnut,C.H., 3rd (1990) Menstrual history as adeterminant of current bone density inyoung athletes. Journal of the AmericanMedical Association 263(4), 545–548.

Edwards, J.E., Lindeman, A.K., Mikesky,A.E. & Stager, J.M. (1993) Energybalance in highly trained femaleendurance runners. Medicine and Science in Sports and Exercise 25(12),1398–1404.

Ellison, P.T. & Lager, C. (1986) Moderaterecreational running is associated withlowered salivary progesterone profilesin women. American Journal of Obstetricsand Gynecology 154, 1000–1003.

Flatt, J.P. (1988). Importance of nutrientbalance in body weight regulation.Diabetes/Metabolism Reviews 4(6),571–581.

Friedl, K E., Moore, R.J., Hoyt, R.W. et al. (2000) Endocrine markers ofsemistarvation in healthy lean men in amultistressor environment. Journal ofApplied Physiology 88(5), 1820–1830.

Frisch, R.E. & McArthur, J.W. (1974)Menstrual cycles: fatness as determinantof minimum weight for height necessaryfor their maintenance or onset. Science185, 949–951.

Gold, P.W., Gwirtsman, H., Averinos, P.C.et al. (1986) Abnormal hypothalamic–pituitary–adrenal function in anorexianervosa: pathophysiologic mechanismsin underweight and weight-correctedpatients. New England Journal of Medicine314(21), 1335–1342.

Goldberg, G.R., Black, A.E., Jebb, S.A. et al. (1991) Critical evaluation of energyintake data using fundamentalprinciples of energy physiology: 1.Derivation of cut-off limits to identifyunder-recording. European Journal ofClinical Nutrition 45(12), 569–581.

Gordon, C.M., Grace, E., Emans, S.J. et al. (2002) Effects of oraldehydroepiandrosterone on bonedensity in young women with anorexianervosa: a randomized trial. Journal ofClinical Endocrinology and Metabolism87(11), 4935–4941.

Grinspoon, S., Thomas, E., Pitts, S. et al.(2000) Prevalence and predictive factorsfor regional osteopenia in women withanorexia nervosa. Annals of InternalMedicine 133(10), 790–794.

Grinspoon, S., Thomas, L., Miller, K.,Herzog, D. & Klibanski, A. (2002) Effectsof recombinant human IGF-I and oral

Page 263: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 247

reversible? Medicine and Science in Sportsand Exercise 35, S12.

Hohmann, J.G., Teal, T.T., Clifton, D.K. et al. (2000) Differential role ofmelanocortins in mediating leptin’seffects on feeding and reproduction.American Journal of Physiology.Regulatory, Integrative and ComparitivePhysiology 278, R50–R59.

Hotta, M., Fukuda, I., Sato, K. et al. (2000)The relationship between bone turnoverand body weight, serum insulin-likegrowth factor (IGF) I, and serum IGF-binding protein levels in patients withanorexia nervosa. Journal of ClinicalEndocrinology and Metabolism 85(1),200–206.

Hubert, P., King, N.A. & Blundell, J.E.(1998) Uncoupling the effects of energyexpenditure and energy intake: appetiteresponse to short-term energy deficitinduced by meal omission and physicalactivity. Appetite 31(1), 9–19.

Ihle, R. & Loucks, A.B. (2004) Dose–response relationships between energyavailability and bone turnover in youngexercising women. Journal of Bone andMineral Research 19, 1231–1240.

Jenkins, A.B., Markovic, T.P., Fleury, A. & Campbell, L.V. (1997) Carbohydrateintake and short-term regulation ofleptin in humans. Diabetologia 40(3),348–351.

Jenkins, P.J., Ibanez-Santos, X., Holly, J. et al. (1993) IGFBP-1: a metabolic signalassociated with exercise-inducedamenorrhoea. Neuroendocrinology 57(4),600–604.

Kaiserauer, S., Snyder, A.C., Sleeper, M. & Zierath, J. (1989) Nutritional,physiological, and menstrual status ofdistance runners. Medicine and Science inSports and Exercise 21(2), 120–125.

Keen, A.D. & Drinkwater, B.L. (1997)Irreversible bone loss in formeramenorrheic athletes. OsteoporosisInternational 7(4), 311–315.

King, N.A., Lluch, A., Stubbs, R.J. &Blundell, J.E. (1997) High dose exercisedoes not increase hunger or energyintake in free living males. EuropeanJournal of Clinical Nutrition 51(7),478–483.

Knobil, E. (1990) The GnRH pulsegenerator. American Journal of Obstetricsand Gynecology 163(5 Pt. 2), 1721–1727.

Kolaczynski, J.W., Considine, R.V.,Ohannesian, J. et al. (1996a) Responses of leptin to short-term fasting andrefeeding in humans: a link withketogenesis but not ketones themselves.Diabetes 45(11), 1511–1515.

et al. (1992) Hypothalamic–pituitary–thyroidal function in eumenorrheic andamenorrheic athletes. Journal of ClinicalEndocrinology and Metabolism 75(2),514–518.

Loucks, A.B., Verdun, M. & Heath, E.M.(1998) Low energy availability, not stressof exercise, alters LH pulsatility inexercising women. Journal of AppliedPhysiology 84(1), 37–46.

McLean, J.A., Barr, S.I. & Prior, J.C. (2001)Cognitive dietary restraint is associatedwith higher urinary cortisol excretion in healthy premenopausal women.American Journal of Clinical Nutrition 73,7–12.

Maddalozzo, G.F. & Snow, C.M. (2000)High intensity resistance training: effects on bone in older men andwomen. Calcified Tissue International66(6), 399–404.

Maffei, M., Halass, J., Ravussin, E. et al.(1995) Leptin levels in human androdent: measurement of plasma leptinand ob RNA in obese and weight-reduced subjects. Nature Medicine 1,1155–1161.

Manning, J.M. & Bronson, F.H. (1989)Effects of prolonged exercise on pubertyand luteinizing hormone secretion in female rats. American Journal ofPhysiology 257(6 Pt. 2), R1359–R1364.

Manning, J.M. & Bronson, F.H. (1991)Suppression of puberty in rats byexercise: effects on hormone levels andreversal with GnRH infusion. AmericanJournal of Physiology 260(4 Pt. 2),R717–R723.

Marcus, R., Cann, C., Madvig, P. et al.(1985) Menstrual function and bonemass in elite women distance runners.Endocrine and metabolic features.Annals of Internal Medicine 102(2),158–163.

Marcus, R., Feldman, D. & Kelsey, J. (1996)Osteoporosis. Academic Press, San Diego,CA.

Mertz, W., Tsui, J.C., Judd, J.T. et al. (1991)What are people really eating? Therelation between energy intake derivedfrom estimated diet records and intakedetermined to maintain body weight.American Journal of Clinical Nutrition54(2), 291–295.

Minami, S., Kamegai, J., Sugihara, H. et al.(1995) Central glucoprivation evoked byadministration of 2-deoxy-D-glucoseinduces expression of the c-fos gene in a subpopulation of neuropeptide Yneurons in the rat hypothalamus. BrainResearch. Molecular Brain Research 33(2),305–310.

Kolaczynski, J.W., Ohannesian, J.P.,Considine, R.V., Marco, C.C. & Caro, J.F.(1996b) Response of leptin to short-termand prolonged overfeeding in humans.Journal of Clinical Endocrinology andMetabolism 81, 4162–4165.

Lane, M.A., Black, A., Handy, A.M. et al.(2001) Energy restriction does not alter bone mineral metabolism orreproductive cycling and hormones in female rhesus monkeys. Journal ofNutrition 131(3), 820–827.

Laughlin, G.A. & Yen, S.S.C. (1996)Nutritional and endocrine-metabolicaberrations in amenorrheic athletes.Journal of Clinical Endocrinology andMetabolism 81(12), 4301–4309.

Laughlin, G.A. & Yen, S.S.C. (1997)Hypoleptinemia in women athletes:absence of a diurnal rhythm withamenorrhea. Journal of ClinicalEndocrinology and Metabolism 82(1),318–321.

Leibel, R.L., Rosenbaum, M. & Hirsch, J.(1995) Changes in energy expenditureresulting from altered body weight. New England Journal of Medicine 332(10),621–628.

Lennkh, C., de Zwaan, M., Bailer, U. et al.(1999) Osteopenia in anorexia nervosa:specific mechanisms of bone loss. Journalof Psychiatric Research 33(4), 349–356.

Levin, B.E., Dunn-Meynell, A.A. & Routh,V.H. (1999) Brain glucose sensing andbody energy homeostasis: role in obesityand diabetes. American Journal ofPhysiology 276(5 Pt. 2), R1223–R1231.

Loucks, A.B. (2004) Energy balance andbody composition in sports and exercise.Journal of Sports Sciences 22, 1–14.

Loucks, A.B. & Horvath, S.M. (1985)Athletic amenorrhea: a review. Medicineand Science in Sports and Exercise 17(1),56–72.

Loucks, A.B. & Thuma, J.R. (2003)Luteinizing hormone pulsatility isdisrupted at a threshold of energyavailability in regularly menstruatingwomen. Journal of Clinical Endocrinologyand Metabolism 88(1), 297–311.

Loucks, A.B., Horvath, S.M. & Freedson,P.S. (1984) Menstrual status andvalidation of body fat prediction inathletes. Human Biology 56(2), 383–392.

Loucks, A.B., Mortola, J.F., Girton, L. &Yen, S.S.C. (1989) Alterations in thehypothalamic–pituitary–ovarian and the hypothalamic–pituitary–adrenalaxes in athletic women. Journal of ClinicalEndocrinology and Metabolism 68(2),402–411.

Loucks, A.B., Laughlin, G.A., Mortola, J.F.

Page 264: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

248 chapter 18

Mizuno, T.M., Kleopoulos, S.P., Bergen,H.T. et al. (1998) Hypothalamic pro-opiomelanocortin mRNA is reduced byfasting and in ob/ob and db/db mice,but is stimulated by leptin. Diabetes47(2), 294–297.

Mizuno, T.M., Makimura, H., Silverstein, J.et al. (1999) Fasting regulateshypothalamic neuropeptide Y, agouti-related peptide, andproopiomelanocortin in diabetic miceindependent of changes in leptin orinsulin. Endocrinology 140(10),4551–4557.

Mulligan, K. & Butterfield, G.E. (1990)Discrepancies between energy intakeand expenditure in physically activewomen. British Journal of Nutrition 64(1),23–36.

Muroya, S., Yada, Y., Shioda, S. &Takigawa, M. (1999) Glucose-sensitiveneurons in the rat arcuate nucleuscontain neuropeptide Y. NeuroscienceLetters 264(1–3), 113–116.

Myerson, M., Gutin, B., Warren, M.P. et al. (1991) Resting metabolic rate andenergy balance in amenorrheic andeumenorrheic runners. Medicine andScience in Sports and Exercise 23, 15–22.

National Collegiate Athletic Association(2003) 2004 NCAA Wrestling Rules andInterpretations. National CollegiateAthletic Association, Indianapolis, IN: WR23–WR35, WI10–WI11,WA9–WA10, WA27–WA32.

Nelson, M.E., Fisher, N.C., Catsos, P.D. et al. (1986) Diet and bone status inamenorrheic runners. American Journal of Clinical Nutrition 43(6), 910–916.

Nielsen, T.F., Ravn, P., Bagger, Y.Z.,Warming, L. & Christiansen, C. (2004)Pulsed estrogen therapy in prevention ofpostmenopausal osteoporosis. A 2-yearrandomized, double blind, placebo-controlled study. OsteoporosisInternational 15(2), 168–174.

Obici, S., Wang, J., Chowdury, R. et al.(2002) Identification of a biochemicallink between energy intake and energyexpenditure. Journal of ClinicalInvestigation 109(12), 1599–1605.

Okano, H., Mizunuma, H., Soda, M. et al. (1995) Effects of exercise andamenorrhea on bone mineral density inteenage runners. Endocrine Journal 42(2),271–276.

Otis, C.L., Drinkwater, B., Johnson, M.,Loucks, A. & Wilmore, J. (1997)American College of Sports Medicineposition stand. The female athlete triad.Medicine and Science in Sports and Exercise29(5), i–ix.

intake in humans after manipulation of glycogen stores. American Journal ofClinical Nutrition 65(4), 941–946.

Soyka, L.A., Grinspoon, S., Levitsky, L.L.,Herzog, D.B. & Klibanski, A. (1999) The effects of anorexia nervosa on bonemetabolism in female adolescents.Journal of Clinical Endocrinology andMetabolism 84(12), 4489–4496.

Soyka, L.A., Misra, M., Frenchman, A. et al.(2002) Abnormal bone mineral accrual inadolescent girls with anorexia nervosa.Journal of Clinical Endocrinology andMetabolism 87(9), 4177–4185.

Stacey, E., Korkia, P., Hukkanen, M.V.J.,Polak, J.M. & Rutherford, O.M. (1998)Decreased nitric oxide levels and boneturnover in amenorrheic athletes withspinal osteopenia. Journal of ClinicalEndocrinology and Metabolism 83(9),3056–3061.

Stefanis, N., Mackintosh, C., Abraha, H.D.,Treasure, J. & Moniz, C. (1998)Dissociation of bone turnover inanorexia nervosa. Annals of ClinicalBiochemistry 35(Pt. 6), 709–716.

Stubbs, R.J., Harbron, C.G., Murgatroyd,P.R. & Prentice, A.M. (1995a) Covertmanipulation of dietary fat and energydensity: effect on substrate flux and food intake in men eating ad libitum.American Journal of Clinical Nutrition62(2), 316–329.

Stubbs, R.J., Ritz, P., Coward, W.A. & Prentice, A.M. (1995b) Covertmanipulation of the ratio of dietary fat tocarbohydrate and energy density: effecton food intake and energy balance infree-living men eating ad libitum.American Journal of Clinical Nutrition62(2), 330–337.

Styne, D.M. & Grumbach, M.M. (1991)Disorders of puberty in the male andfemale. In: Reproductive Endocrinology(Yen, S.S.C. & Jaffe, R.B., eds.). W.B.Saunders, Co., Philadelphia, PA:511–544.

Tabata, I., Ogita, F., Miyachi, M. &Shibayama, H. (1991) Effect of low bloodglucose on plasm CRF, ACTH, andcortisol during prolonged physicalexercise. Journal of Applied Physiology 71,1807–1812.

Thong, F.S., McLean, C. & Graham, T.E.(2000) Plasma leptin in female athletes:relationship with body fat, reproductive,nutritional, and endocrine factors.Journal of Applied Physiology 88(6),2037–2044.

Trabulsi, J. & Schoeller, D.A. (2001)Evaluation of dietary assessmentinstruments against doubly labeled

Pirke, K.M., Schweiger, U., Broocks, A.,Tuschl, R.J. & Laessle, R.G. (1990)Luteinizing hormone and folliclestimulating hormone secretion patternsin female athletes with and withoutmenstrual disturbances. ClinicalEndocrinology 33, 345–353.

Ravussin, E. (2002) Cellular sensors of feast and famine. Journal of ClinicalInvestigation 109(12), 1537–1540.

Rivier, C. & Rivest, S. (1991) Effect of stresson the activity of the hypothalamic–pituitary–gonadal axis: peripheral and central mechanisms. Biology ofReproduction 45, 523–532.

Rossetti, L. (2000) Perspective:hexosamines and nutrient sensing.Endocrinology 141(6), 1922–1925.

Sanborn, C.F., Albrecht, B.H. & Wagner,W.W., Jr. (1987) Athletic amenorrhea:lack of association with body fat.Medicine and Science in Sports and Exercise19(3), 207–212.

Schneider, J.E. & Wade, G.N. (2000)Inhibition of reproduction in service of energy balance. In: Reproduction in Context: Social and EnvironmentalInfluences on Reproductive Physiology andBehavior (Wallen, K. & Schneider, J.E.,eds.). MIT Press, Cambridge, MA:35–82.

Schwartz, M.W., Sipols, A.J., Marks, J.L. et al. (1992) Inhibition of hypothalamicneuropeptide Y gene expression by insulin. Endocrinology 130(6), 3608–3616.

Scott, E.C. & Johnston, F.E. (1982) Criticalfat, menarche, and the maintenance of menstrual cycles: a critical review.Journal of Adolescent Health Care 2(4),249–260.

Selye, H. (1939) The effect of adaptation tovarious damaging agents on the femalesex organs in the rat. Endocrinology 25,615–624.

Sinning, W.E. & Little, K.D. (1987) Bodycomposition and menstrual function in athletes. Sports Medicine 4(1), 34–45.

Sjodin, A.M., Andersson, A.B., Hogberg,J.M. & Westerterp, K.R. (1994) Energybalance in cross-country skiers: a studyusing doubly labeled water. Medicine and Science in Sports and Exercise 26(6),720–724.

Slentz, C.A., Davis, J.M., Settles, D.L., Pate, R.R. & Settles, S.J. (1990) Glucosefeedings and exercise in rats: glycogenuse, hormone responses, andperformance. Journal of AppliedPhysiology 69, 989–994.

Snitker, S., Larson, D.E., Tataranni, P.A. & Ravussin, E. (1997) Ad libitum food

Page 265: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

influence of energy availability 249

water, a biomarker of habitual energyintake. American Journal of Physiology.Endocrinology and Metabolism 281(5),E891–E899.

Trappe, T.A., Gastaldelli, A., Jozsi, A.C.,Troup, J.P. & Wolfe, R.R. (1997) Energyexpenditure of swimmers during highvolume training. Medicine and Science inSports and Exercise 29(7), 950–954.

Treloar, A.E., Boynton, R.E., Behn, B.G. & Brown, B.W. (1967) Variation of the human menstrual cycle throughreproductive life. International Journal ofFertility 12(1 Pt. 2), 77–126.

Veldhuis, J.D., Evans, W.S., Demers, L.M.et al. (1985) Altered neuroendocrineregulation of gonadotropin secretion inwomen distance runners. Journal ofClinical Endocrinology and Metabolism61(3), 557–563.

Wade, G.N. & Jones, J.E. (2003) Lessonsfrom experimental disruption of estrous cycles and behaviors. Medicine and Science in Sports and Exercise 35,1573–1580.

Wade, G.N. & Schneider, J.E. (1992)Metabolic fuels and reproduction in female mammals. Neuroscience and Biobehavioral Reviews 16(2), 235–272.

Wade, G.N., Schneider, J.E. & Li, H.Y.(1996) Control of fertility by metaboliccues. American Journal of Physiology270(1 Pt. 1), E1–E19.

Wang, J., Liu, R., Hawkins, M., Barzilai, N.& Rossetti, L. (1998) A nutrient-sensingpathway regulates leptin geneexpression in muscle and fat. Nature393(6686), 684–688.

in the induction of menstrual cycledisturbances during strenuous exercisetraining. Journal of Clinical Endocrinologyand Metabolism 86(11), 5184–5193.

Wilmore, J.H., Wambsgans, K.C., Brenner,M. et al. (1992) Is there energyconservation in amenorrheic comparedwith eumenorrheic distance runners?Journal of Applied Physiology 72(1), 15–22.

Yahiro, J., Glass, A.R., Fears, W.B.,Ferguson, E.W. & Vigersky, R.A. (1987)Exaggerated gonadotropin response toluteinizing hormone-releasing hormonein amenorrheic runners. AmericanJournal of Obstetrics and Gynecology156(3), 586–591.

Zanker, C.L. & Swaine, I.L. (1998a) Bone turnover in amenorrhoeic andeumenorrhoeic women distancerunners. Scandinavian Journal of Medicineand Science in Sports 8(1), 20–26.

Zanker, C.L. & Swaine, I.L. (1998b)Relation between bone turnover,oestradiol, and energy balance inwomen distance runners. British Journalof Sports Medicine 32(2), 167–171.

Zanker, C.L. & Swaine, I.L. (1998c) Therelationship between serum oestradiolconcentration and energy balance inyoung women distance runners.International Journal of Sports Medicine19(2), 104–108.

Zanker, C.L., Cooke, C.B., Truscott, J.G.,Oldroyd, B. & Jacobs, H.S. (2004) Annualchanges of bone density over 12 years inan amenorrheic athlete. Medicine andScience in Sports and Exercise 36(1),137–142.

Warren, M.P., Brooks-Gunn, J., Fox, R.P. et al. (2003) Persistent osteopenia inballet dancers with amenorrhea anddelayed menarche despite hormonetherapy: a longitudinal study. Fertilityand Sterility 80(2), 398–404.

Weigle, D.S., Duell, P.B., Connor, W.E. et al. (1997) Effect of fasting, refeeding,and dietary fat restriction on plasmaleptin levels. Journal of ClinicalEndocrinology and Metabolism 82,561–565.

Westerterp, K.R. & Saris, W.H. (1991)Limits of energy turnover in relation tophysical performance, achievement ofenergy balance on a daily basis. Journal of Sports Sciences 9, 1–15.

Westerterp, K.R., Meijer, G.A., Janssen,E.M., Saris, W.H. & Ten Hoor, F. (1992)Long-term effect of physical activity onenergy balance and body composition.British Journal of Nutrition 68(1), 21–30.

Williams, N.I., Young, J.C., McArthur, J.W.et al. (1995) Strenuous exercise withcaloric restriction: effect on luteinizinghormone secretion. Medicine and Sciencein Sports and Exercise 27(10), 1390–1398.

Williams, N.I., Caston-Balderrama, A.L.,Helmreich, D.L. et al. (2001a)Longitudinal changes in reproductivehormones and menstrual cyclicity incynomolgus monkeys during strenuousexercise training: abrupt transition toexercise-induced amenorrhea.Endocrinology 142(6), 2381–2389.

Williams, N.I., Helmreich, D.L., Parfitt,D.B., Caston-Balderrama, A.L. &Cameron, J.L. (2001b) Evidence for acausal role of low energy availability

Page 266: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

250

Oral contraceptive use is common in both recre-ational and elite female athletes. Oral contraceptivesare generally comprised of a combination of estro-gen- and progesterone-like compounds. The com-position is dependant upon the varying levels of the two steroid derivatives produced by the variousmanufacturers. In addition to their use for contra-ception, women may also be prescribed oral contra-ceptives for cycle regulation and to manage variouscycle dysfunctions (i.e. amenorrhea or dysmenor-rhea). Side effects such as weight gain, fluid retentionand nausea initially caused many athletic women to avoid the use of oral contraceptives. However,with the introduction of lower dose formulationsthe negative side effects have been reduced and thenumbers of women using these drugs has increased.The impact of the oral administration of ovariansteroids on physical performance has been evalu-ated over the past several decades. Research, how-ever, remains incomplete as to the potential positiveand/or negative effects of these compounds on sportperformance. The following chapter will addressthe endogenous hormonal control of the menstrualcycle, the various types and mechanisms of action of exogenous hormones and the influence of thesedrugs on physical performance.

Overview of normal menstrual cycle

The menstrual cycle is regulated primarily by agroup of five hormones; gonadotropin-releasinghormone (GnRH), follicle-stimulating hormone(FSH), luteinizing hormone (LH), estrogen and pro-gesterone. These hormones are released as part of a

classical endocrine axis known as the hypothalamic–pituitary–gonadal (HPG) axis (Fig. 19.1). Regulationof the HPG axis occurs through both short- andlong-loop feedback systems.

GnRH is secreted from the hypothalamus in apulsatile manner throughout the menstrual cycle.Hypothalamic GnRH pulsatility is essential for regu-lar menstrual cyclicity. On average, the frequency ofGnRH secretion is once per 90 min during the earlyfollicular phase and once per 60–70 min during theluteal phase. The release of both FSH and LH isinduced by GnRH with LH being the more sensit-ive of the two hormones with respect to changes inGnRH levels (Larsen et al. 2003).

FSH is secreted by the anterior pituitary glandand is essential for follicular growth. Its secretion is highest and most critical during the 1st week of the follicular stage. At the level of the ovary, FSHinduces estrogen and progesterone secretion byactivating aromatase and p450 enzymes. FSH alsoinduces the proliferation of granulosa cells andexpression of LH receptors on granulosa cells.

LH is secreted by the anterior pituitary gland andis required for both growth of preovulatory folliclesand luteinization and ovulation of the dominant follicle. During the follicular phase of the menstrualcycle, LH induces androgen synthesis by theca cells;stimulates proliferation, differentiation, and secre-tion of follicular thecal cells; and increases LHreceptors on granulosa cells. The preovulatory LHsurge drives the oocyte into the first meiotic divisionand initiates luteinization of thecal and granulosacells. The resulting corpus luteum produces highlevels of progesterone and some estrogen.

Chapter 19

Oral Contraceptive Use and Physical Performance

JACI L. VANHEEST, CARRIE E. MAHONEY AND CAROL D. RODGERS

Page 267: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

oral contraceptive use and physical performance 251

Estrogen is produced at the level of the ovary and is crucial for the development of the antrumand maturation of the Graafian follicle. Estrogen ispredominant at the end of the follicular phase,directly preceding ovulation. Estradiol is primarilyderived from androgens produced by thecal cells.The androgens migrate from the thecal cells to thegranulosa cells, where they are converted into estra-diol by aromatase enzyme. The actions of estradiolinclude induction of FSH receptors on granulosacells, proliferation and secretion of follicular thecalcells, induction of LH receptors on granulosa cells,and proliferation of endometrial stromal and epi-thelial cells. At low circulating levels, estrogens

exert negative feedback on LH and FSH secretion;however, at very high levels estrogens exert positivefeedback on LH and FSH secretion. Estrogen furtherinduces proliferation of granulosa cells and syn-thesis of estrogen receptors, establishing a positivefeedback loop on itself. In the uterine endometrialcycle, estrogen induces proliferation of the endo-metrial glands (Knobil 1999).

In the presence of a GnRH pulse, the pituitary and ovarian hormones exert mutual control over thecirculating levels of each other. The complex inter-actions between pituitary and ovarian hormonesinvolve forward control, positive feedback, andnegative feedback mechanisms. They also serve tosustain a self-perpetuating monthly endocrine cycle.Figure 19.2 illustrates the relationships between therelative amounts of key hormones of the menstrualcycle. Day one of the menstrual cycle begins withmenstruation, which occurs at the beginning of thefollicular phase.

The follicular phase of the menstrual cycle spansthe first day of menstruation until ovulation. Theprimary purpose of the follicular phase is to developa viable follicle capable of undergoing ovulation.The early events of the follicular phase are initiatedby a rise in FSH levels that occurs on the 1st day of the cycle. This rise in FSH levels can be attributedto a decrease in progesterone and estrogen levels at the end of the previous cycle and the subsequ-ent removal of their inhibitory effect on FSH. FSHstimulates the development of 15–20 follicles eachmonth and stimulates follicular secretion of estra-diol by up-regulating secretion of androgens by the theca externa, and by inducing the aromataseenzyme receptor on granulosa cells (Yen 1999). FSHfurther induces expression of FSH receptors by fol-licles. As estradiol levels increase under the influ-ence of FSH, estradiol inhibits the secretion of FSHand FSH levels decrease.

Under normal circumstances, one follicle evolvesinto the dominant follicle, destined for ovulation,while the remaining follicles undergo atresia. It iscurrently not known how the dominant follicle isselected; yet it has been observed that the dominantfollicle always expresses an abundance of FSHreceptors. As FSH levels decrease towards the endof the follicular phase, the developing follicles must

Hypothalamus

LHRH

FSH

Ovaries

Pituitary

LH

EstrogenProgesterone

Fig. 19.1 The hypothalamic–pituitary–gonadal (HPG)axis. FSH, follicle-stimulating hormone; LH, luteinizinghormone; LHRH, luteinizing hormone-releasinghormone.

Page 268: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

252 chapter 19

compete for relatively small amounts of FSH. Thedominant follicle, with its high concentration ofFSH receptors is able to acquire more FSH even as FSH levels decrease. This enables the dominantfollicle to continue to synthesize estradiol which is essential for its complete maturation. Since theremaining follicles can no longer produce theneeded amount of estradiol due to the decreasing

FSH level they cease to develop and ultimatelyundergo atresia. The dominant follicle may alsorelease paracrine factors that stimulate apoptosis in the other follicles. As the dominant follicle con-tinues to mature it secretes increasing amounts of estrogen. Estrogen levels peak towards the end ofthe follicular phase of the menstrual cycle. At thiscritical point, estrogen exerts positive feedback on

Hypothalamus

Control by hypothalamus1)

2)

3)

4)

Releasing hormone

FSH

FSH

LH

FSH LH

LH

Inhibited by combination ofestrogen and progesterones

Stimulated by high levelsof estrogen

Anterior pituitary

Pituitary hormones in blood

Ovarian cycle

LH peak triggersovulation andcorpus luteumformation

Growing follicle Maturingfollicle

OvulationCorpusluteum

Degeneratingcorpus luteum

Post-ovulatory phasePre-ovulatory phase

Estrogen

Estrogen

0

Estrogen

Progesteroneand estrogen

Progesterone

Progesteroneand estrogen

Ovarian hormones in blood

5) Menstrual cycle

Menstruation Days

Endometrium

5 10 1514 20 25 28

Fig. 19.2 The relationships betweenthe relative amounts of key hormonesof the menstrual cycle. FSH, follicle-stimulating hormone; LH, luteinizinghormone.

Page 269: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

oral contraceptive use and physical performance 253

LH (positive feedback is usually associated withhaving estradiol present in excess of 1100 pmol·L–1

for > 24 h), generating a dramatic preovulatory LHsurge. It is important to note that estrogen can onlyexert a positive feedback on LH at this precise stage in the menstrual cycle; if estrogen is artificiallyprovided earlier in the cycle, ovulation will not beinduced (Yen 1999; Larsen et al. 2003).

The luteal phase is characterized by the luteiniza-tion of those components of the follicle that were not ovulated. It is initiated by the LH surge. Thegranulosa cells, theca cells and some surroundingconnective tissue are all converted into the corpusluteum, which eventually undergoes atresia. Themajor effects of the LH surge are the conversion of granulosa cells from predominantly androgen-converting cells to predominantly progesterone-synthesizing cells by the expression of new LHreceptors. This fosters increased progesterone syn-thesis, and a reduced affinity of granulose cells forestrogen and FSH in these cells. Combined, thesechanges promote increased progesterone secretionwith some estrogen secretion. Progesterone secre-tion by the corpus luteum peaks between 5 and 7days post-ovulation. High progesterone levels exertnegative feedback on GnRH and subsequentlyGnRH pulse frequency decreases. As GnRH pulsefrequency decreases, FSH and LH secretion alsodecreases. The corpus luteum further loses its FSHand LH receptors. The lack of FSH and LH stimula-tion precipitates atresia of the corpus luteum and its subsequent evolution into the corpus albicans.With the decline of both estrogen and progesteronelevels, an important negative feedback control onFSH is removed and FSH levels rise once again toinitiate the next menstrual cycle (Yen 1999).

The LH surge is required for ovulation. Under theinfluence of LH, the primary oocyte enters the finalstage of the first meiotic division and divides into asecondary oocyte and the first Barr body. The LHsurge induces release of proteolytic enzymes, whichdegrade the cells at the surface of the follicle, andstimulates angiogenesis in the follicular wall andprostaglandin secretion. These effects of LH causethe follicle to swell and rupture. At ovulation, theoocyte is expelled into the peritoneal cavity. Theoocyte adheres to the ovary and muscular contrac-

tions of the fallopian tube bring the oocyte into con-tact with the tubal epithelium to initiate migrationthrough the oviduct.

Generally, estrogen (estradiol) is produced con-tinuously during the cycle. It is very low during the early follicular phase, rising to a peak during the late follicular phase and triggering ovulation.Progesterone and estrogen increase during theluteal phase, and the increased estrogen causes the endometrium, the inner lining of the uterus, tothicken and mature. Progesterone helps to matureand stabilize the endometrium. It also prevents further endometrial proliferation and mitosis andchanges the endometrium to a secretory structurethat is ready for implantation of the fertilized ovum.At the end of the luteal phase, concentrations ofestrogen and progesterone decrease, which triggersmenstruation and the subsequent breakdown of the endometrium. Breakdown of the endometriumoccurs assuming no implantation of an ovum hasoccurred during this period.

Inhibin is a glycoprotein that has been isolated as a heterodimer with two subunits linked by dis-ulphide bonds. The two isoforms of inhibin aretermed inhibin A and inhibin B. Inhibin is producedby the granulosa cells and the corpus luteum infemales (deKrester & Robertson 1989; Groome et al.1996). Control of inhibin synthesis and release is viathe hormones FSH (granulosa cells) and LH (corpusluteum). Level of inhibin (serum) peak mid-cycleand in the mid-luteal phase, with a decline in con-centrations prior to menstruation (Sehested et al.2000). Inhibin has FSH-suppressing properties, aswell as, more recently discovered immune, nervoussystem and hemopoietic properties (deKrester &Robertson 1989; Sehested et al. 2000). In addition,inhibin may have paracrine actions related togrowth and maturation of the ovary and classicalendocrine actions in the maturation of the HPG axis(Groome et al. 1996; Sehsted et al. 2000). Additionalresearch is necessary to fully elucidate the role ofinhibin in human physiology.

Overview of oral contraceptives

Oral contraceptive pills are available in three majorformulationsafixed-dose, combination phasic and

Page 270: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

254 chapter 19

daily progestin. Monophasic pill formulations con-tain a fixed dose of estrogen and progestin through-out the cycle. Biphasic and triphasic (multiphasic)pills reduce the total amount of progestin through-out the cycle as to mimic normal physiologic levels.Ethinyl estradiol and mestranol are typical syntheticestrogens used in oral contraceptive formulas. Olderformulations contained mestranol in high doses (50 µg) compared to the newer low dose formula-tions (< 50 µg of estrogen compounds ethinyl estra-diol or mestranol). First generation progestins in-clude norethindrone and norethindrone acetate orethynodiol diacetate and norethynodrel. Secondgeneration progestins are typically norgesterel andlevonorgestrel (Larsen et al. 2003). Finally, desoges-trel, norestimate and gestrodene are third genera-tion progestins. The third generation progestins aredesigned to be selective for progestin receptors,thus reducing the atherogenic properties of thesecompounds (Godsland et al. 1992). Table 19.1 listsoral contraceptives that are currently available inthe USA. It is important to note that the new genera-tion progestins are assumed to be less androgeniccompared to the early formulations. However, allcombination oral contraceptives are beneficial in theclinical treatment of various conditions includinghirsuitism (Thorneycroft 1999). Low dose oral con-traceptives are considered to have a lipid neutralaffect resulting in a reduced atherosclerosis rates inhumans.

First generation oral contraceptives were associ-ated with many negative side effects such as weightgain, fatigue, headaches and nausea (Lebrun et al.2003). These side effects have been minimized bythe second and third generation lower dose drugs.These formulations have 30–40% lower levels ofhormones resulting in significantly reduced sideeffects (Greenblatt 1985). Studies evaluating changesin body weight and body composition in both non-active and athletic women using oral contraceptivessupport these findings. Female athletes (Lebrun et al. 2003) and non-athletes (Rosenberg 1998) reportnon-significant weight gains (~ 1 kg over 6 weeks) or no change in body weight and body composi-tion, respectively. However, the impact of smallbody weight and body composition changes asso-ciated with oral contraceptive use in female athletes

cannot be overlooked when discussing elite sport performance.

Mechanism of action of oralcontraceptives

The mechanism by which estrogen and progesteroneprevent ovulation is through the suppression of theFSH and LH mid-cycle surge. Combination formu-lations are highly effective at inhibiting the secretionof gonadotrophic hormone, and through this mech-anism prevent ovulation. Conversely, progestin-only products are inconsistent in the suppression ofovulation and operate through the inhibition of therelease of GnRH from the hypothalamus. In thisway, progestin only products attenuate the levels ofFSH, LH, estradial and progesterone (Speroff et al.1993).

These induced changes in circulating hormonelevels precipitate changes in endometrial function-ing. Changes in ovum transport and implantation,coupled with alterations in the composition of thecervical mucus (increase viscosity and reduce vol-ume) are also evident (Larsen et al. 2003). Together,the hormonal and endometrial alterations result in a negative environment for success of ovulation,implantation and support of the ovum.

Oral contraceptives also impact carbohydrate,protein and fat metabolism (Table 19.2). Carbohy-drate metabolism is influenced by the dose, potencyand chemical structure of the progestin in the drug. The role of the synthetic estrogens on glucosemetabolism is unclear, however they may act syn-ergistically with the progestins to cause impairedglucose tolerance. Epidemiological studies indicatelow risk or no risk for developing diabetes in oralcontraceptive users (new generation). However, it isprudent to consider the glucose handling status ofeach woman when prescribing oral contraceptives.Low dose norethindrone-type progestins are prefer-able in these women.

Estrogens used in oral contraceptives increase thehepatic production of various globulins such asFactor V, VII, X and fibrinogen. These globulinsenhance thrombosis. In addition, synthetic estro-gens also increase the synthesis of angiotensinogen(which is converted to angiotensin) resulting in elev-

Page 271: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

oral contraceptive use and physical performance 255

ated blood pressure in some women (Larsen 2003).The androgenic progestins increase the synthesis ofsex hormone binding globulin (SHBG). SHBG bindsthe 19-nortestosterone compounds used in oral con-traceptives. Overall, the incidence of venous andarterial thrombosis is directly related to the dose of the estrogen, which is relatively low in the newgeneration formulations.

Synthetic estrogens used in oral contraceptivescause an increase in high-density lipoproteins (HDL),

total cholesterol and triglycerides, and a decrease in low-density lipoproteins (LDL) (Larsen 2003).Conversely, progestin elements cause a decrease inHDL, total cholesterol and triglycerides, and anincrease in LDL. The negative impact of the pro-gestins in the early oral contraceptives was relatedto the high androgenic activity of these compounds.Today’s low dose oral contraceptives are less an-drogenic and appear to have a more lipid neutralresponse (Thorneycroft 1999). When evaluating the

Table 19.1 Oral contraceptive formulations. (Data from Mishell 1999 and Larsen et al. 2003.)

Product Brand name type Progestagen Estrogen

Levlen C 0.15 mg levonorgestrel 30 µg ethinylestradiolTri-Levlen C, T 0.5 or 0.075 or 0.125 mg levonorgestrel 30 µg/40 µg /30 µg ethinylestradiolOvcon-35 C 0.4 mg norethindrone 35 µg ethinylestradiolOvcon-50 C 1.0 mg levonorgestrel 50 µg ethinylestradiolDesogen C 0.15 mg desogestrel 35 µg ethinylestradiolMircette B, C 0.15 mg/0 mg desogestrel 30 µg/10 µg ethinylestradiolMicronor P 0.35 mg norethindroneModicon C 0.5 mg norethindrone 35 µg ethinylestradiolOrtho-Cept C 0.15 mg desogestrel 30 µg ethinylestradiolOrtho-Cyclen C 0.25 mg norgestimate 35 µg ethinylestradiolOrtho-Novum 1/35 C 1.0 mg norethindrone 35 µg ethinylestradiolOrtho-Novum 1/50 C 1.0 mg norethindrone 50 µg mestranolOrtho-Novum C, T 0.5 mg/0.75 mg/L mg norethindrone 35 µg ethinylestradiolOrtho-Novum C, B 0.5 mg/1.0 mg norethindrone 35 µg ethinylestradiolOrtho-tricyclin C, T 0.18 mg/0.215 mg/0.25 mg norgestimate 35 µg ethinylestradiolEstrostep C, T 1.0 mg norethindrone 20 µg/30 µg/35 µg ethinylestradiolLoestrin 1/20 C 1.0 mg norethindrone 20 µg ethinylestradiolLoestrin 1.5/30 C 1.5 mg norethindrone 30 µg ethinylestradiolNorlestrin 1/50 C 1.0 mg norethindrone 50 µg ethinylestradiolNorlestrin 2.5/50 C 2.5 mg norethindrone 50 µg ethinylestradiolBrevicon C 0.5 mg norethindrone 35 µg ethinylestradiolNorinyl 1 + 35 C 1.0 mg norethindrone 35 µg ethinylestradiolNorinyl 1 + 50 C 1.0 mg norethindrone 50 µg ethinylestradiolNor-Q.D. P 0.35 mg norethindroneTri-Norinyl C, T 0.5 mg/1 mg/0.5 mg norethindrone 35 µg ethinylestradiolDemulen 1/35 C 1.0 mg ethynodiol diacetate 35 µg ethinylestradiolDemulen 1/50 C 1.0 mg ethynodiol diacetate 50 µg ethinylestradiolAlesse C 0.1 mg levonorgestrel 20 µg ethinylestradiolLo/Ovral C 0.3 mg levonorgestrel 30 µg ethinylestradiolNordette C 0.15 mg levonorgestrel 30 µg ethinylestradiolOvral C 0.5 mg norgestrel 50 µg ethinylestradiolOvrette P 75 mg norgestrel 30 µg ethinylestradiolTriphasil C, T 50 mg/75 mg/125 mg levonorgestrel 40 µg /40 µg /30 µg ethinylestradiol

B, biphasic; C, combination; P, progestagen only; T, triphasic.Androgenic activity (relative to 1 mg of norethindrone: norethindrone (1 mg) = 1; levonorgestrel (1 mg) = 8.3;drospirenone (1 mg) = 0; desogestrel (1 mg) = 3.4; norgestimate (1 mg) = 1.9; ethynodiol diacetate (1 mg) = 0.6).

Page 272: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

256 chapter 19

metabolic response to various oral contraceptives, itis critical that the type, dose and combination ofeach synthetic hormone be assessed.

Impact of oral contraceptive use onphysical performance

The literature regarding the impact of oral contra-ceptive use on performance has grown substantiallyover the past several decades. Early studies explor-ing the effect of first generation, high dose drugs areoften distinctly different from the more recent workevaluating the low dose multiphasic compounds.While the focus of this review will be on the secondand third generation drugs that are currently beingused by women participating in sport, earlier workwill be discussed when appropriate.

Aerobic performance

Maximal oxygen consumption (Vo2max) is an oftenused measure when assessing aerobic ability.Normal ovarian hormone cyclicity does not appearto affect Vo2max (De Souza et al. 1990; Bemben et al.1992, 1995; Lebrun et al. 1995; Lynch & Nimmo1998); however, data examining the impact of hor-monal alterations as induced by changes in exogen-ous hormones, such as would occur with oralcontraceptive use, is less conclusive.

Studies evaluating the administration of exogen-ous ovarian hormones have varied in the dose and

type of oral contraceptives utilized. Research in the early 1980s failed to demonstrate any significantdifference in performance between oral contracept-ive users and non-users (McNeill & Mozingo 1981;Huisveld et al. 1983). A subsequent short duration(21 day) evaluation of 1 mg norethindrone also did not demonstrate a significant difference inVo2max between oral contraceptive users and non-users (Bryner et al. 1996). Interestingly, however,endurance trained women who used monophasicoral contraceptives for a 6-month period experi-enced a significant decrease in Vo2peak (Notelovitz et al. 1987). Further evaluation of a triphasic pre-paration performed by Casazza et al. (2002) withmoderately active women also showed a significantreduction (–11%) in Vo2peak following 4 months oforal contraceptive use. Peak heart rate and minuteventilation, however, were not different Theseresults were supported by the work of Lebrun et al. (2003) who showed a 4.7% decrease in Vo2maxfollowing a 2-month triphasic oral contraceptiveintervention protocol.

Potential factors that could impact maximal aerobic capacity include stroke volume, oxygentransport capacity, oxygen extraction capacity andmuscle blood flow. The reductions in Vo2max maylogically be associated with a reduction in strokevolume and/or oxygen transport capacity. Strokevolume has been shown to increase in perimeno-pausal women following hormone replacementtherapy (Kamali et al. 2000). However, resting

Table 19.2 Metabolic effects of oral contraceptives. (Data from Dorflinger 1985 and Godsland et al. 1992.)

Steroid derivative Substrate pathway Metabolic effect

Ethinyl estradiol Protein Decrease amino acidsEthinyl estradiol Carbohydrate No change in plasma insulin or glucose toleranceEthinyl estradiol Lipid Increase cholesterol, HDL and triglyceride

Decrease LDL cholesterol19-Nortestosterone derivatives Proteins None19-Nortestosterone derivatives Carbohydrate Increase plasma insulin

Decrease glucose tolerance19-Nortestosterone derivatives Lipid Decrease cholesterol, HDL and triglyceride

Increase LDL cholesterol

HDL, high-density lipoprotein; LDL, low-density lipoprotein.

Page 273: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

oral contraceptive use and physical performance 257

ferritin, hemoglobin concentration and serum ironconcentration remain unchanged in women usingoral contraceptives (Mooij et al. 1992) which wouldsuggest that these primary factors are not the mech-anism by which aerobic potential is limited.

Blood flow regulation via mechanisms such assympathetic nervous system (SNS) activation hasalso been cited as a factor that may be responsiblefor the observed reductions in maximal aerobiccapacity. The evaluation of various conditions whereovarian hormones are elevated provides experi-mental evidence for this mechanism. In pregnancywhere there is a period of high estrogen and proges-terone concentrations SNS activation is suppressed,in conjunction with a decreased level of circulatingcatecholamines (McMurray et al. 1993). In this way,a reduction in SNS activation with oral contracept-ive administration might also be responsible for thedecreases in aerobic capacity shown in these studies.

Anaerobic and strength performance and muscle damage

The current literature on the relationship of exo-genous administration of ovarian hormones andanaerobic metabolism and/or muscular strength isextremely limited. Early work by Petrofsky et al.(1976) identified a reduction in muscle strength during the luteal phase of normally cycling females.The potential for oral contraceptive use to blunt this strength decline was subsequently evaluated ina group of athletic women by Lebrun et al. (2003).Anaerobic performance on the anaerobic speed testand isokinetic strength measured on the Cybexdynamometer was determined prior to and follow-ing administration of a triphasic formulation for 2 months. Although this study failed to find a sig-nificant difference in either anaerobic performanceor isokinetic strength, the relationship of high levelsof ovarian hormones to anaerobic and/or strengthperformance capacity is significantly underexam-ined and must be further researched before anydefinitive conclusions can be established.

The influence of endogenous and/or exogenousestrogens on exercise induced muscle damage hasbeen evaluated over the past decade. Ultrastruc-

tural muscle damage is a typical consequence ofstrenuous exercise such as resistance training. Therole of estradiol on membrane structure and per-meability has been evaluated in both animal andhuman models.

Women have responded to strenuous exercisewith lower serum creatine kinase (CK) concentra-tions compared to men, which has been associatedwith lower total muscle mass in women (Shumate et al. 1979; Rogers et al. 1985). However, animal studiessupport the role of estradiol in reducing membranepermeability in response to exercise (Amelink et al.1988, 1990; Bar & Amelink 1997).

Research evaluating the role of estradiol on muscle damage following exercise in women usingoral contraceptives is both limited and equivocal.Miles and Schneider (1993) reported no influence of estradiol on CK activity following exercise. Therelationship between delayed onset muscle sore-ness and oral contraceptive use was reported fol-lowing a 50-min stepping routine in women usingoral contraceptives compared to controls. Oral con-traceptives were associated with a reduction in self-reported soreness scores; however, the mechanismfor these findings remains unclear (Thompson et al.1997).

Recent studies have utilized eccentric muscledamage protocols. Downhill running (30 min) wasused by Carter et al. (2001) to elicit damage in oralcontraceptive users and non-users. Both groupsexperienced significantly elevated CK and musclesoreness scores compared to baseline measures.Differences were evident between the groups at 72 h following damage with the oral contraceptiveusers exhibiting a reduction in CK compared to thecontrol group. These data support a protective roleof estrogen following muscle damage. In contrast,Savage and Clarkson (2002), using a 50 repetitioneccentric muscle contraction protocol of the elbowflexors, showed no difference in measures of sore-ness, serum CK or range of motion of the elbowjoint. However, oral contraceptive users had delayedforce recovery (maximal isometric strength) at 2days following muscle damage.

The role of exogenous estrogens on muscle damage following exercise remains unclear. Animal

Page 274: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

258 chapter 19

studies support the protective role (anti-oxidantcharacteristics) of estradiol on membrane perme-ability. Human studies vary in methodology andoutcome measures. Further research is necessary toclarify the potential benefits of estrogen on musclestability following strenuous physical activity.

Heat tolerance

Menstrual cycle phase is associated with variedresponses to thermal stressors. Exercise during theluteal phase is associated with an elevated core tem-perature (0.4°C) compared to the follicular phase(Stachenfeld et al. 2000). In contrast, oral contracept-ive users have less variability in core temperaturebetween phases than non-users. The primary factorfor these phasic alterations in core temperature isthe oscillations in progesterone across the men-strual cycle (elevation during the luteal phase). Useof oral contraceptives containing progestins wouldcause elevations in core temperature during exer-cise similar to women during the luteal phase. Theinfluence of progesterone on core temperature isfurther supported by the response of women usingthe long-term contraceptive method of injectableprogesterone (i.e. Depot-Provera [Cheung et al.2000]). These women experience an elevated coretemperature during the 24–36 h following admin-istration of the drug. Martin and Buono (1997)reported elevated core temperature (0.3°C) andheart rate (8 b·min–1) in women using oral contra-ceptives containing synthetic progestins while exer-cising in the heat (30°C, 50% relative humidity).These changes in core temperature and heart rateare similar to those seen in women exercising in theheat during the luteal phase. It appears that secondand third generation oral contraceptives mimic thethermoregulatory responses of women exercising ina hot environment during the luteal phase. Furtherresearch is necessary to more clearly understand theimpact of low dose contraceptive use on perform-ance in hot and humid environments.

Substrate utilization and metabolic flux

The metabolic effects of contraceptive steroids varied depending on the formulation. Table 19.2

illustrates the influence of estrogen ethinyl estradioland the 19-nortestosterone derivative progrestinson metabolism. The influence of the oral contra-ceptives is directly related to dosage and potency of the steroid derivative in the formulation. Themetabolic effects at rest have been evaluated in relation to side effects of the drugs. Only recentlyhave scientists begun to understand the role of oralcontraceptive use in female athletes during physicalexercise.

Current research on the metabolic alterationsassociated with oral contraceptive use during exercise is limited. Cross-sectional studies havecompared oral contraceptive users with non-users.Early work by Bonen et al. (1991) reported signi-ficantly elevated free fatty acid concentrations in oral contraceptive users when compared withnon-users during basal or resting conditions. This tendency also appears to occur during exercise with reductions in respiratory exchange ratio (RER)and a potential enhancement in lipid oxidation also evident in oral contraceptive users (Bemben et al.1995).

More recently, studies using stable isotope tech-nology to evaluate both glucose and lipid metabol-ism during exercise and at rest have demonstrated a decrease in glucose rate of appearance (Ra), dis-appearance rate (Rd) and metabolic clearance rate in women using oral contraceptive during exerciseon a cycle ergometer (45% or 65% Vo2peak) (Suh et al.2003). Under similar exercise conditions, triglycer-ide mobilization was measured using glycerol rateof appearance (infusion of [1,1,2,3,3-2H]glycerol).Glycerol Ra increased significantly at both exerciseintensities in the oral contraceptive condition. Theelevated Ra was associated with a significantlyincreased plasma cortisol concentration but no dif-ference in RER (Casazza et al. 2004). These datawould suggest that endogenous ovarian hormonesdo play a role in affecting glucose and lipid meta-bolism during exercise. The exercise induced hor-monal perturbations clearly dictate the metabolicflux in normal cycling women. Oral contraceptives(triphasic) provide a greater stimulus to lipid andglucose flux during exercise conditions. The level ofcirculating ovarian hormones appears to be criticalin these metabolic alterations.

Page 275: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

oral contraceptive use and physical performance 259

Summary

Oral contraceptive use has grown in females parti-cipating in sport and physical activity. The role thatthese exogenous ovarian hormones play in physicalperformance remains to be fully elucidated. It isclear that third generation lower dose formulationsresult in reduced side effects for women. Thesechanges have made the use of oral contraceptivesmore appealing to a broad group of female athletesincluding elite caliber performers.

The reductions seen in aerobic capacity appear to be associated with potential alterations in bloodflow due to suppression of SNS activation coupledwith a reduction in circulating catecholamines. Thesefindings are similar to those seen during pregnancy.Anaerobic and/or strength performance appear to be unchanged during oral contraceptive use.

Potential thermoregulatory issues exist for womenusing oral contraceptives; however, the responsesmimic women exercising during the luteal phase ofthe menstrual cycle. The most prominent findingregarding oral contraceptive use during exercise is on substrate metabolism. Studies using stable isotope methodologies have provided significantinsight into the interplay between lipid and glucosemetabolism during exercise in women using exogen-ous hormones.

The literature examining oral contraceptive useduring exercise is relatively limited. It is importantto consider the potential negative or ergogeniceffects of exogenous ovarian steroid use in elitefemale athletes. Longitudinal studies of longer dura-tions are necessary to provide practical recommenda-tions to female athletes.

References

Amelink, G.J., Kamp, H.H. & Bar, P.R.(1988) Creatine kinase isoenzymeprofiles after exercise in the rat: sex-linked differences in leakage of CK-MM.Pflugers Archiv 412, 417–421.

Amelink, G.J., Koot, R.W., Erich, W.B.M.,Van Gijn, J. & Bar, P.R. (1990) Sex-linkedvariation in creatine kinase release, andits dependence on oestradiol, can bedemonstrated in an in-vitro rat skeletalmuscle preparation. Acta PhysiologicaScandinavica 138, 115–124.

Bar, P.R. & Amelink, G.J. (1997) Protectionagainst muscle damage exerted byoestrogen: hormonal or antioxidantaction? Biochemical Society Transactions25, 50–54.

Bemben, D.A., Boileau, R.A., Bahr, J.M.,Nelson, R.A. & Misner, J.E. (1992) Effectsof oral contraceptives on hormonal andmetabolic responses during exercise.Medicine and Science in Sports and Exercise24, 434–441.

Bemben, D.A., Salm, P.C. & Salm, A.J.(1995) Ventilatory and blood lactateresponses to maximal treadmill exerciseduring the menstrual cycle. Journal ofSports Medicine and Physical Fitness 35,257–262.

Bonen, A., Haynes, F.J. & Graham, T.E.(1991) Substrate and hormonalresponses to exercise in women usingoral contraceptives. Journal of AppliedPhysiology 70, 1917–1927.

Bryner, R.W., Toffle, R.C., Ullrich, I.H. &Yeater, R.A. (1996) Effect of low doseoral contraceptives on exerciseperformance. British Journal of SportsMedicine 30, 36–40.

Casazza, G.A., Suh, S.H., Miller, B.F.,Navazio, F.M. & Brooks, G.A. (2002)Effects of oral contraceptives on peakexercise capacity. Journal of AppliedPhysiology 93, 1698–1702.

Casazza, G.A., Jacobs, K.A., Suh, S.H. et al.(2004) Menstrual cycle phase and oralcontraceptive effects on triglyceridemobilization during exercise. Journal ofApplied Physiology 97(1), 302–309.

Carter, A., Dobridge, J. & Hackney, A.C.(2001) Influence of estrogen on markersof muscle tissue damage followingeccentric exercise. Fiziologiia Cheloveka27, 133–137.

Cheung, S.S., McLellan, T.M. & Tenaglia, S.(2000) The thermophysiology ofuncompensatable heat stress.Physiological manipulations andindividual characteristics. SportsMedicine (Auckland, N.Z.) 29, 329–359.

deKretser, D.M. & Robertson, D.M. (1989) The isolation and physiology of inhibin and related proteins. Biology of Reproduction 40, 33–47.

De Souza, M.J., Maguire, M.S., Rubin, K.R.& Maresh, C.M. (1990) Effects ofmenstrual phase and amenorrhea onexercise performance in runners.

Medicine and Science in Sports and Exercise22(5), 575–580.

Dorflinger, L. (1985) Relative potency ofprogestins used in oral contraceptives.Contraception 557, 31–37.

Godsland, I.F., Crook, D. & Wynn, V.(1992) Clinical and metabolicconsiderations of long-term oralcontraceptive use. American Journal ofObstetrics and Gynecology 166, 1955–1964.

Greenblaat, R.B. (1985) Oralcontraceptives: the state of the art.Clinical Therapeutics 8, 6–27.

Groome, N.P., Illingworth, P.J., O’Brien,M. et al. (1996) Measurement of dimericinhibin B throughout the humanmenstrual cycle. Journal of ClinicalEndocrinology and Metabolism 81,1401–1405.

Huisveld, I.A., Haspers, J.E.H. & Bernink, M.J. (1983) The effect of oralcontraceptives and exercise onhemostatic and fibrinolytic mechanismsin trained women. International Journal ofSports Medicine 4, 97–103.

Kamali, P., Muller, T., Lang, U. & Clapp, J.F., 3rd. (2000) Cardiovascularresponses of perimenopausal women tohormone replacement therapy. AmericanJournal of Obstetrics and Gynecology 182,17–22.

Knobil, E. (1999) The wisdom of the bodyrevisited. News in Physiological Sciences14, 1–11.

Page 276: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

260 chapter 19

Larsen, P.R., Kronenberg, H.M., Melmed,S. & Polonsky, K.S. (2003) WilliamsTextbook of Endocrinology. W.B. Saunders,Co., Philadelphia, PA.

Lebrun, C.M., McKenzie, D.C., Prior, J.C. &Taunton, J.E. (1995) Effects of menstrualcycle phase on athletic performance.Medicine and Science in Sports and Exercise27, 437–444.

Lebrun, C.M., Petit, M.A., McKenzie, D.C.,Taunton, J.E. & Prior, J.C. (2003)Decreased maximal aerobic capacitywith use of a triphasic oral contraceptivein highly active women: a randomizedcontrolled trial. British Journal of SportsMedicine 37, 315–320.

Lynch, N.J. & Nimmo, M.A. (1998) Effects of menstrual cycle and oralcontraceptive use on intermittentexercise. European Journal of AppliedPhysiology 78, 565–572.

McMurray, R.G., Mottola, M.F., Wolfe,L.A. et al. (1993) Recent advances inunderstanding maternal and fetalresponses to exercise. Medicine andScience in Sports and Exercise 25,1305–1321.

McNeill, A.W. & Mozingo, E. (1981)Changes in the metabolic cost ofstandardized work associated with the use of an oral contraceptive. Journal of Sports Medicine 21, 238–244.

Martin, J.G. & Buono, M.J. (1997) Oralcontraceptives elevate core temperatureand heart rate during exercise in theheat. Clinical Physiology (Oxford, England)17, 401–409.

Miles, M.P. & Schneider, C.M. (1993)Creatine kinase isoenzyme MB may beelevated in healthy young women aftersubmaximal eccentric exercise. Journal

adolescent girls and adult women:relation to age, stage of puberty,menstrual cycle, follicle-stimulatinghormone, luteinizing hormone, andestradiol levels. Journal of ClinicalEndocrinology and Metabolism 85,1634–1640.

Shumate, J.B., Brooke, M.H., Carroll, J.E. & Davis, J.E. (1979) Increased serumcreatine kinase after exercise: a sex-linked phenomenon. Neurology 29,902–904.

Speroff, L., DeCherney, A. & the AdvisoryBoard for the New Progestins (1993)Evaluation of the new generation of oralcontraceptives. Obstetrics and Gynecology81, 1034–1044.

Stachenfeld, N.S., Silva, C. & Keefe, D.L. (2000) Estrogen modifies thetemperature effects of progesterone.Journal of Applied Physiology 88, 1643–1649.

Suh, S.H., Casazza, G.A., Horning, M.A.,Miller, B.F. & Brooks, G.A. (2003) Effectsof oral contraceptives on glucose fluxand substrate oxidation rates during restand exercise. Journal of Applied Physiology94, 285–294.

Thompson, H.S., Hyatt, J.P., De Souza, M.J. & Clarkson, P.M. (1997) The effectsof oral contraceptives on delayed onsetmuscle soreness following exercise.Contraception 56, 59–65.

Thorneycroft, I.H. (1999) Update onandrogenicity. American Journal of Obstetrics and Gynecology 180, 288–294.

Yen, S.S.C. (1999) The human menstrualcycle: neuroendocrine regulation. In:Reproductive Endocrinology: Physiology,Pathophysiology and Clinical Management(Yen, S.S.C., Jaffe, R.B. & Barbieri, R.L.,eds.). W.B. Saunders, Co., Philadelphia,PA: 191–217.

of Laboratory Clinical Medicine 122,197–201.

Mishell, D.R. (1999) Contraception. In:Reproductive Endocrinology: Physiology,Pathophysiology and Clinical Management(Yen, S.S.C., Jaffe, R.B. & Barbieri, R.L.,eds.). W.B. Saunders, Co., Philadelphia,PA: 676–708.

Mooij, P.N.M., Thomas, C.M.G., Doesburg,W.H. & Eskes, T.K.A.B. (1992) Theeffects of oral contraceptives andmultivitamin supplementation on serumferritin and hematological parameters.International Journal of ClinicalPharmacology and Therapeutics 30, 57–62.

Notelovitz, M., Zauner, C. & McKenzie, I.(1987) The effect of low-dosecontraceptives on cardiorespiratoryfunction, coagulation, and lipids inexercising young women: a preliminaryreport. American Journal of Obstetrics andGynecology 156, 591–598.

Petrofsky, J.S., LeDonne, D.M. & Rinehart,J.S. (1976) Isometric strength andendurance during the menstrual cycle.European Journal of Applied Physiology 35,1–10.

Rogers, M.A., Stull, G.A. & Apple, F.S.(1985) Creatine kinase isoenzymeactivities in men and women following a marathon race. Medicine and Science in Sports and Exercise 17, 679–682.

Rosenberg, M. (1998) Weight change withoral contraceptive use and during themenstrual cycle. Contraception 58, 45–49.

Savage, K.J. & Clarkson, P.M. (2002) Oralcontraceptive use and exercise-inducedmuscle damage and recovery.Contraception 66, 67–71.

Sehested, A., Juul, A., Andersson, A.M. et al. (2000) Serum inhibin A and inhibinB in healthy prepubertal, pubertal and

Page 277: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

261

Introduction

Historically, opportunities for women to engage inphysical activity increased dramatically with thepassing of Title IX in the USA. To date, over threedecades of subsequent research examining theimpact of exercise on women’s bodies and beha-vioral aspects of the athletic lifestyle have broughtattention to gender-specific issues such as muscu-loskeletal health, weight and diet concerns, and theimpact of exercise on the menstrual cycle. Lookingback, it appears that research on the musculoskel-etal effects of exercise and weight and diet concernsdeveloped soon after observations of disturbancesin reproductive function associated with exercise.After an early report documenting a higher pre-valence of menstrual abnormalities in athletes than non-athletes (Erdelyi 1962), numerous otherstudies confirmed this finding in the late 1970s andearly 1980s (Feicht et al. 1978; Dale et al. 1979; Bakeret al. 1981), focusing mostly on amenorrhea anddelayed menarche in long-distance runners (Feichtet al. 1978) and ballet dancers (Frisch et al. 1980;Warren 1980). Interestingly, while Frisch and Revelle(1971) had put forth the hypothesis that menarchedepended on the achievement of a critical bodyweight level of body fat, Warren (1980) observedthat changes in body weight and body compositiondid not correlate with the onset of menarche indancers who acutely decreased their exercise due toinjury. Warren thus popularized the concept thatthe ‘energy drain’ or metabolic cost of exercise

might be a contributing factor to alterations in menstrual cyclicity in female athletes. The first links between exercise-associated amenorrhea andcompromised bone were made by Drinkwater et al.(1984) and Cann et al. (1984) who observed signific-antly lower bone mineral content in amenorrheicrunners. Drinkwater et al. (1990) then documented a significant relationship between lumbar spinebone mineral density (BMD) and menstrual historyin female athletes. An authoritative review byLoucks and Horvath in 1985 (Loucks & Horvath1985) helped highlight the importance of exercise-associated menstrual disturbances (EAMD) as aresearch area and synthesized the available informa-tion concerning plausible mechanisms of the inter-actions between exercise and reproductive function.Researchers’ attention was focused on the potentialcontributions of body composition, training habits,diet, physical stress and psychological stress in theetiology of EAMD. In 1987, a link between ‘eatingproblems’ and amenorrhea in ballet dancers wasreported by Brooks-Gunn et al. (1987), and in 1991,Wilmore (1991) put forth the idea that athletes inboth endurance or appearance sports are at anincreased risk for disordered eating, secondaryamenorrhea and bone mineral disorders. Shortlyafter, the female athlete triad (Fig. 20.1), i.e. a con-dition comprised of restrictive eating, menstrualdisorders and skeletal demineralization was recog-nized (Yeager et al. 1993), and subsequently describedin a Position Stand published by the AmericanCollege of Sports Medicine (ACSM) in 1997 (Otis

Chapter 20

Energy Balance and Exercise-AssociatedMenstrual Cycle Disturbances: Practical andClinical Considerations

NANCY I. WILLIAMS AND MARY JANE DE SOUZA

Page 278: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

262 chapter 20

et al. 1997). Since that time, recognition of the import-ance of the triad and the need to more specificallydefine the components, establish the prevalence,physiological underpinnings and the clinical con-sequences has driven the research in the area ofexercise and the menstrual cycle, and stimulated arevision of the 1997 ASCM Position Stand. A currentunderstanding of the practical and clinical consid-erations of the effects of exercise on the menstrualcycle in today’s active woman must include thebroader scope of the female athlete triad, and con-sequently recognize the complexity of interrelation-ships among its components. A multidisciplinaryapproach to this issue involving behavioral, physio-logical, sociocultural and medical perspectives iswarranted. This chapter will provide current infor-mation on the practical issues relating to the impactof exercise on menstrual function, such as theidentification, prevention and treatment of men-strual disorders, an exploration of factors that pre-dispose certain individuals to menstrual disordersand considerations for the restoration of normalmenstrual function. Clinical issues arising from theassociation of physical activity and disturbances inreproductive function will also be examined. Theseinclude the impact of endocrine changes associ-ated with menstrual disturbances on fertility, bonehealth and possibly the cardiovascular system, andthe association of menstrual disturbances withrestrictive eating.

Exercise-associated menstrualdisturbances: definitions and prevalence

Overview

Important aspects of the relationship between exer-cise and the menstrual cycle have been revealedthrough descriptive, observational, cross-sectionaland prospective studies utilizing both survey methods and physiological approaches. Our currentunderstanding is that EAMD exist on a spectrum ofseverity, i.e. luteal phase defects (LPD), anovulationand amenorrhea. Prior to discussing the definitionsand prevalence of the different EAMD, attention is warranted toward the appropriate use of terms.The term ‘exercise-induced menstrual disturbances’implies that menstrual disturbances are directlyrelated to participation in or the stress of physicalexercise per se. Contrary to this idea, a variety ofexperimental approaches have yielded evidencethat inadequate caloric intake is a causal factor inmenstrual disturbances with exercise (Laughlin &Yen 1996; Loucks et al. 1998; Williams 1998; Williamset al. 2001b; De Souza et al. 2003) such that a state oflow energy availability created by an imbalance ofenergy consumed as food and expended throughexercise is the key factor in the onset of reproductivesuppression. The use of a more fitting term is thuswarranted, i.e. ‘exercise-associated’ or ‘exercise-related’ menstrual disturbances. Since estimates of

Disorderedeating

Exercise-associatedmenstrual disorders

Bone lossEndothelialdysfunction?

Psychologicalstress

Is energy deficiency the only factor contributing to EAMD?

Energydeficiency

?

Thefemaleathletetriad

Fig. 20.1 The relationship betweencomponents of the female athletetriad. Disordered eating andexercise combined to produce low energy availability, causingexercise-associated menstrualdisturbances (EAMD). Psychosocialstress is theoretically depicted as afactor that may exacerbate theeffects of low energy availability on EAMD. (Modified and reprintedwith permission from Williams2003.)

Page 279: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 263

the prevalence of secondary amenorrhea are higherin athletes than in sedentary populations (Drew1961), there is a correlation between athletic amenor-rhea and physical exercise, but there is not a causalrelationship.

Amenorrhea

Figure 20.2 depicts this continuum of menstrual disturbances ranging from subtle perturbations likeLPD and anovulatory cycles to the most severe pre-sentation, amenorrhea (De Souza 2003). In athletes,the amenorrhea is hypothalamic in origin and ismanifested by suppressed levels of gonadotropinsand ovarian steroids (Loucks & Horvath 1985;Velduis et al. 1985). The definition of amenorrhea inthe literature has varied, but should be conservat-ively defined as no menses for a minimum of 3months due to the clinical sequelae associated withchronic estrogen deficiency (Loucks & Horvath1985). The prevalence of amenorrhea in athletesranges from 1% to 66% (Feicht et al. 1978; Dale et al.1979; Schwartz et al. 1981; Sanborn et al. 1982, 2000;Loucks & Horvath 1985), is highest in those sportswith an aesthetic component like gymnastics andfigure skating, and grossly exceeds estimates of thiscondition in sedentary women (2–5%) (Drew 1961;Petterson et al. 1973).

Delayed menarche

Delayed menarche, or primary amenorrhea, defined

as failure to achieve menarche by the age of 16 years(Loucks & Horvath 1985), has been repeatedlyreported in athletes participating in many sports,but again particularly in the aesthetic sports. Thelater ages at menarche in adolescent athletes areoften attributed to regular exercise training withoutconsidering other factors known to influence pub-ertal growth and maturation (Malina 1994). Malina(1994) states that in adequately nourished adoles-cents, the timing of menarche is very dependent onhereditary factors; but menarche is also influencedby a number of social or biocultural variables,including the self-selective nature of participation in some sports, like gymnastics, figure skating andballet, where selection occurs for specific factorsassociated with a delayed or later maturation(Malina 1994). The mechanisms that link these fac-tors to a later age of maturation remain undefined.Following the review of several studies that exam-ined menstrual disturbances in athletes, the averageage of menarche was about 1 year later (13 vs. 12 years old) in the groups of amenorrheic athletes,but these differences were not statistically significantin the individual studies. In both human (Warren1980) and animal studies (Cheung et al. 1997), how-ever, the timing of puberty has been related to nutri-tional status (Wade et al. 1996), and specifically, toincreased leptin concentrations providing a per-missive effect on growth (Cheung et al. 1997). Sinceleptin concentrations can discriminate athletes ofdiffering menstrual status (Laughlin & Yen 1997; DeSouza et al. 2003), and evidence exists for a causal

Continuum of menstrual disturbances in athletes

Anovulat

ion

Oligom

enorrh

ea

Amen

orrhea

Lute

al phas

e def

ect

Ovulat

ory

Fig. 20.2 Continuum of reproductive disturbances, ranging from ovulatory cycles, subtle presentations of luteal phasedefects (LPD) and anovulatory cycles to the most severe menstrual disturbance, amenorrhea. Physically active womenand athletes fluctuate between ovulatory cycles, LPD and anovulatory disturbances frequently. It also seems probablethat amenorrheic athletes may experience LPD during recovery from amenorrhea. (Reprinted with permission from DeSouza 2003.)

Page 280: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

264 chapter 20

role of energy availability in the development ofmenstrual disturbances (Williams et al. 2001a), it isreasonable to speculate that energy status may playa role in the common observance of a later age ofmenarche in amenorrheic athletes, if participationin exercise began prior to puberty.

Oligomenorrhea

Oligomenorrhea is defined by irregular and incon-sistent menstrual cycles lasting from 36–90 days inlength (Loucks & Horvath 1985). The key parameterof interest in the definition of oligomenorrhea is the unpredictable nature of the intervals betweenmenstrual cycles. Given these inconsistent charac-teristics, it is a menstrual presentation that is diffi-cult to study. As such, no definitive data exist on the prevalence of oligomenorrhea in athletes, exceptto note that cycles of irregular length are oftenreported in female athletes (Loucks & Horvath1985) and oligomenorrhea as a menstrual categoryis frequently grouped together with amenorrhea inmany studies (Gremion et al. 2001; Csermely et al.2002; Cobb et al. 2003). When daily measurement of hormones has not been feasible or affordable,investigators have also used definitions that haveincluded 3–4 or fewer menstrual cycles per year todefine oligomenorrhea (Cobb et al. 2003). The ovar-ian profile of an oligomenorrheic athlete displayserratic, unpredictable and presumably inadequateestradiol (E2) production as a given follicle strugglesto achieve dominance, but certainly may result in anovulatory cycle in an unpredictable manner.

Anovulation

Anovulation is defined as the absence of ovulationof an oocyte in the face of inadequate luteinizinghormone (LH) secretion secondary to inadequateestrogen priming in the follicular phase and theobvious absence of luteinization (Hamilton-Fairly & Taylor 2003). Anovulatory cycles are character-ized by low E2 and progesterone (P4) levels through-out the cycle; however, much debate in the clinicalforum continues regarding the specific criterion for confirming anovulation (Malcolm & Cumming2003). Because serial ultrasound measurements to

document ovulation are not always feasible, manyresearchers have relied on daily assessment of urinary ovarian steroid metabolites and urinary LHto confirm ovulation (or anovulation). Using thisapproach, De Souza et al. (1998a) have reported a16% prevalence of anovulatory cycles in womenthat exercise at recreational levels despite havingcharacteristic regular menstrual intervals of 26–32days. Williams et al. (2000) have reported that in 32%of Division 1 athletes from a wide variety of sportswho self-reported regular menstrual bleeding of26–32 days, ovulation could not be detected. Inmost cases, a higher degree of estrogen exposure ispresent in anovulatory cycles compared to amenor-rheic cycles; that is an anovulatory cycle is likely tohave greater E2 production in a 30-day period thanan equivalent period in an amenorrheic athlete. Inboth of the latter conditions, the absence of proges-terone from luteinized cells renders estrogen actionson some tissues unopposed.

LPD have been reported in women engaged in alllevels of physical activity, from strenuous to recre-ational exercise, (Shangold et al. 1979; Ellison &Lager 1986; Broocks et al. 1990; Beitins et al. 1991;Winters et al. 1996; De Souza et al. 1998a; De Souza2003). In women with LPD, the ovarian system func-tions at a level adequate for ovulation, but inade-quate to support successful implantation, since the latter is dependent on adequate exposure of theendometrium to P4 (Balash & Vanrell 1987). Areduction in luteal phase P4 production and abbre-viated luteal phases are the key characteristics ofLPD. Reduced P4 production during the lutealphase is also referred to as luteal phase inadequacyor insufficiency to describe the poor quality of theendometrium secondary to the low P4 levels. Thelow P4 levels that occur are either low in volume orlow in duration of output, since they occur in theface of normal menstrual cycle lengths of 26–32days ( Jones 1976; Balash & Vanrell 1987). The otherpresentation of LPD in athletes is the shortening ofthe luteal phase, referring to luteal phases of 10 daysor less (Sherman & Korenman 1974; Jones 1976; DeSouza 2003). Clinically, LPD-associated P4 inade-quacy causes asynchronous follicular growth in thesubsequent menstrual cycle, compromised oocytematuration and differentiated (out-of-phase) func-

Page 281: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 265

tion of the endometrium. All of the latter factors are associated with low rates of cycle fecundity andhigh rates of embryonic loss, i.e. infertility andspontaneous abortion (Jones 1976; Balash & Vanrell1987). It is important to understand that womenwith exercise-associated LPD continue to ovulate,although some women do so as late as day 20 (DeSouza 2003), reflective of the short luteal phases thatare apparent in association with some presentationsof LPD. The prevalence of LPD in non-active (i.e.sedentary) women is controversial, but estimatesvary from 2% to 5%, and from 3% to 20% in womenwith infertility (Balash & Vanrell 1987; McNeely &Soules 1988). LPD occur in athletes at a muchgreater prevalence of approximately 79% than isreported in non-active women, representing themost common menstrual cycle abnormality associ-ated with exercise (De Souza 1998a). It is importantto note that EAMD occurs outside the competitiveathletic arena as even recreationally active womenhave been recognized to exhibit a high prevalence ofLPD (De Souza et al. 1998a). For a thorough reviewof LPD in athletes, see a recent review by De Souza(2003).

Exercise-associated menstrualdisturbances: clinical considerations

Infertility

A clinical consequence of exercise-associated amen-orrhea and anovulatory cycles is temporary infer-tility. This represents an obvious problem forwomen attempting to conceive. With amenorrhea,the absence of menses is noticeable and thus theindividual is aware that conception is unlikely.However, if there is a spontaneous resumption ofovulation, pregnancy is possible because ovula-tion may not be preceded by a return of menses.Anovulatory cycles can be accompanied with regu-lar and consistent menstrual bleeding intervals and thus the individual is not aware that conceptionis unlikely. This is in sharp contrast to irregularcycle intervals indicative of oligomenorrheic cycleswhere ovulation is unpredictable. Although diffi-cult to diagnose, LPD may be the most commonabnormality observed in athletes and contribute to

10% of infertility and 25% of habitual abortion; how-ever, the infertility is related to poor P4 productionrather than ovulatory problems (Soules 1988). Inwomen desiring to become pregnant, decreases inthe duration or amount of P4 secreted during theluteal phase has been correlated with the low cyclefecundity (Jones & Madigal-Castro 1970; Strott et al.1970). Blacker et al. (1997) found that only very smalldifferences in luteal phase P4 accounted for un-explained infertility in a group of women comparedto aged-matched controls. No differences in existedin the number of preovulatory follicles, the rate offollicular growth, or the mean diameter before fol-licle rupture and timed endometrial biopsy (Blacker et al. 1997). Alternatively, no differences were foundin salivary P4 obtained during the days prior toimplantation in cycles that resulted in conceptionand the levels on the same days that did not result inconception. The major determinant of cycle fecun-dity was the robustness of the follicular phase, as theprobability of conception was significantly relatedto the value of the mid-follicular E2 concentration(Lipson & Ellison 1996). The previous studies sug-gest that subtle changes in luteal function and/or follicular E2 levels may have implications for fertility. To date, no studies have been performed infemale athletes with LPD to assess fertility butclearly the high prevalence of LPD in recreationallyactive women and athletes warrants more work inthis area.

Effects on the cardiovascular system

Due to an observed cardioprotective effect of estrogens, persistently low E2 levels in amenorrheicathletes may also have adverse effects on cardio-vascular health. Clinically, one of the earliest signsof cardiovascular disease is a decrease in endothe-lial function, and is evident decades before overtcoronary artery disease is present (Celermajer 1997;Luscher & Barton 1997; Schachinger et al. 2000).Atherosclerotic disease progression and adversecardiovascular events have both been shown to be associated with a decrease in endothelial func-tion (Celermajer 1997; Luscher & Barton 1997;Schachinger et al. 2000). In addition, physicalchanges to the endothelium and the availability of

Page 282: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

266 chapter 20

nitric oxideaan important endothelial-derived relax-ing factoraare also decreased in association withatherosclerotic disease (Luscher & Barton 1997).

It is well-established that estrogen plays a sig-nificant role in endothelial-dependent blood flowvia nitric oxide and has effects on cholesterol andlipoproteins (Steinberg 1987; Celermajer 1997;Luscher & Barton 1997; Mendelsohn & Karas 1999;Schachinger et al. 2000). As such, hypoestrogenismhas been associated with endothelium-dependentdysfunction (Celermajer 1997; Luscher & Barton1997; Schachinger et al. 2000). The mechanism ofaction of estrogen on endothelial function is likelythrough the nitric oxide pathway, which is knownto be critical in vascular control and during reactivehyperemia (Armour & Ralston 1998; Arora et al.1998; Ayres et al. 1998). Genomic and non-genomiceffects of estrogen have been shown to play asignificant role in the up-regulation of endothelialnitric oxide synthase (eNOS) and the subsequentproduction and increased half-life of nitric oxide(Armour & Ralston 1998; Simoncini et al. 2002). Somestudies (Armour & Ralston 1998; Simoncini et al.2002) have suggested that estrogen affects vascularendothelial release of nitric oxide through actionsthat enhance the bioavailability of nitric oxide (production) by up-regulating the constitutive nitricoxide synthase and, conversely, by inhibiting super-oxide anion production.

Coincident with declining estrogen levels, a studyby Celermajer et al. (1994) has shown a reduction in endothelial-dependent vasodilation as soon as 3 months after natural menopause. Impaired endo-thelial function has also been demonstrated 1 weekafter surgical menopause (Ohmichi et al. 2003). Giventhese rapid reductions in endothelial-dependentvasodilation following both surgical and naturalmenopause, it is logical to question the impact ofclinical and subclinical levels of hypoestrogenism inphysically active women.

Recent data from our laboratory has identifiedreduced peripheral blood flow, and in another laboratory impaired endothelial cell dysfunction inamenorrheic athletes has been reported (Zeni-Hochet al. 2003). These cardiovascular findings are mostlikely the result of chronic hypoestrogenism. Thus,

these results suggest that chronic hypoestrogenismmay predispose young physically active and ath-letic women to early or premature cardiovascular dis-ease. To date, only two studies have examined theimpact of athletic amenorrhea on endothelial function.

Zeni-Hoch et al. (2003) examined brachial arteryflow-mediated dilation (endothelium-dependent) inamenorrheic athletes and compared them to womenwith oligomenorrhea and age-matched controls.Zeni-Hoch et al. (2003) reported that endothelialfunction was 80% lower in athletes with amenor-rhea, compared to athletes with either normal menstrual cycles or oligomenorrhea. Disturbingly,the magnitude of impaired endothelium-dependentvasodilation in amenorrheic athletes was compar-able to data previously reported in otherwise healthypost-menopausal women (Blumel et al. 2003) andolder (60 + 2 years) coronary-artery-disease patients(Celermajer et al. 1992) after a similar flow-mediatedstimulus. In contrast, endothelium-independentdilation to nitroglycerine was not different amongstthe groups. Recent data from our laboratory demon-strate that amenorrheic athletes had lower resting(2.2 + 0.1 vs. 4.8 + 0.4; p < 0.001) and peak ischemic(42.8 + 2.1 vs. 52.9 + 2.0; p = 0.004) blood flow res-ponses (mL·100 mL–1·min–1) using lower limb strain-gauge plethysmography (O’Donnell et al. 2004). Theamenorrheic athletes also had lower resting supineheart rate (50.5 + 4.8 vs. 58.8 + 1.9; p = 0.07) andsupine resting systolic blood pressure (90.4 + 5.7 vs. 106.8 + 2.0 mmHg; p = 0.004), compared to theireumenorrheic counterparts. In light of the knownlink between estrogen and vascular function, theobserved attenuated blood flow response is likely a consequence of the chronic hypoestrogenism inthe amenorrehic athletes. These findings confirmaltered flow-mediated endothelial-dependent vaso-dilation. Lower resting heart rate and systolic bloodpressure may be indicative of altered autonomicregulation, similar to that seen in anorexia nervosapatients. These findings represent an additionalhealth paradigm associated with the female athletetriad and suggests that hypoestrogenism in ame-norrheic athletes may lead to deleterious cardio-vascular outcomes. Thus, the clinical sequelae ofhypoestrogenism in young amenorrheic women

Page 283: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 267

must be extended to include detrimental effects oncardiovascular function that could have seriousconsequences later in life. Since cardiovascularhealth is of concern in this young cohort, more evaluation is warranted.

Bone health

Chronic hypoestrogenism is a major cause of boneloss in women, regardless of age (Ott 1990; Matkovicet al. 1994). It is well-established that the chronichypoestrogenism coincident with menopause is amajor cause of osteoporosis in women (Matkovic et al. 1994; Kanis 2002). Like the anorexic patient,female athletes who experience amenorrhea also ex-perience chronic hypoestrogenism (Zipfel et al. 2001).

Hypoestrogenism associated with secondaryamenorrhea in athletes contributes greatly to boneloss (Cann et al. 1984; Drinkwater et al. 1984, 1990;Marcus et al. 1985; Matkovic et al. 1994). Since men-strual status is a reflection of E2 production, themost severe menstrual perturbations exhibit thegreatest reductions in E2 production, and are asso-ciated with the most severe consequences to bone.The failure to achieve peak bone mass or prematurebone loss, and appropriate clinical diagnostic cri-teria as measured by dual energy X-ray absorptio-metry (DXA), i.e. the use of t-scores, should beencouraged (Ott 1990; Kanis 2002; Khan et al. 2002).Reduced BMD in amenorrheic athletes has beenrepeatedly reported by many investigators (Cann et al. 1984; Drinkwater et al. 1984, 1990; Marcus et al.1985; Keen & Drinkwater 1997; Tomten et al. 1998;Gremion et al. 2001; Csermely et al. 2002; Cobb et al.2003). Most often a t-score of –1.0 to –2.5 is observedin athletes with amenorrhea, and is referred to asosteopenia (Kanis 2002; Khan et al. 2002). The clin-ical criterion of osteopenia is associated with a 100%increase in the risk of fracture and is notably of long-term concern for the management of bone health inthese individuals. The clinical sequelae of a lowpeak bone mass or premature bone loss includes anincreased risk of stress fractures and fractures of thehip and spine (Carbon et al. 1990; Otis et al. 1997;Korpelainen 2001). Moreover, if the clinical criterionof osteoporosis is achieved in an athlete, an even

greater risk of fracture is apparent (Kanis 2002).Similar data are available in women with E2 defici-ency associated with anorexia nervosa. Since expos-ure to E2, along with genetic and nutritional factors,determines peak bone mass (Matkovic et al. 1994;Kanis 2002), a delay in menses related to exercisetraining may result in a lower than normal peakvalue. Peak bone mass appears to be a very goodpredictor of the rate of post-menopausal bone loss,and higher levels will delay the risk of fracture (Ott1990; Matkovic et al. 1994; Kanis 2002).

It is well-documented that amenorrheic athletesunequivocally suffer from reductions in BMD, particularly in the lumbar spine (Cann et al. 1984;Drinkwater et al. 1984, 1990; Marcus et al. 1985; Keen& Drinkwater 1997; Tomten et al. 1998; Gremion et al. 2001; Csermely et al. 2002; Cobb et al. 2003).Even in oligomenorrheic athletes, very low BMDscores have been repeatedly reported (Tomten et al.1998; Gremion et al. 2001; Csermely et al. 2002; Cobbet al. 2003). In one study of oligomenorrheic athletes,lumbar BMD was only 69% of that observed in an aged-matched menstruating group of women(Cobb et al. 2003). The low BMD experienced byamenorrheic athletes is both severe and likely irre-versible, since resumption of menses offers minimalimprovements in BMD, permanently compromis-ing the attainment of peak BMD (Drinkwater et al.1986; Jonnavithula et al. 1993; Keen & Drinkwater1997). Even the administration of oral contracept-ives and other hormonal replacement strategies failto significantly improve BMD in amenorrheic andoligomenorrheic athletes (Cumming 1996; Fagan1998). Dugowson et al. (1991) contend that the boneloss observed in amenorrheic athletes may be seri-ous enough to result in osteoporotic fractures wellbefore menopause.

Based on available data, the prevalence ofosteopenia in amenorrheic athletes is estimated torange from 1.4% to 50% in athletes and exercisingwomen (Drinkwater et al. 1984, 1990; Dugowson et al. 1991; Rutherford 1993; Young et al. 1994;Lauder et al. 1999; Pettersson et al. 1999; Khan et al.2002; Cobb et al. 2003). It is important to recognizethat the clinical endpoint of osteopenia is not neces-sarily accompanied by the other two components of

Page 284: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

268 chapter 20

the triad (disordered eating and amenorrhea). Thatis, the existence of even one of these problems hasbeen associated with a decrease in BMD, presumablybecause both disorders are associated with varyingdegrees of hypoestrogenism (Cobb et al. 2003). Sixpercent of the oligomenorrheic and/or amenorrheicrunners in the study by Cobb et al. (2003) had BMDt-scores that were osteoporotic, whereas 48% hadBMD t-scores that were osteopenic. Cobb et al.(2003) also demonstrated that 26% of the womenwho were menstruating and had some evidence of disordered eating had BMD t-scores that wereosteopenic. Sports medicine professionals shouldaggressively move toward the understanding thatthe existence of even one subclinical entity asso-ciated with the female athlete triad may have a negative clinical impact on the long-term health ofthese athletes and physically active women.

Although physical activity has generally beenconsidered to be protective of BMD, some researchsuggests that a proportion of physically activewomen, even in the face of apparently normal men-strual cycles, have reduced BMD (Prior et al. 1990).Data reported by Prior et al. (1990) suggested thatLPD and anovulatory cycles resulted in progress-ively more spinal bone loss over a 1-year period inmoderate- and long-distance runners, and theyattributed this loss to lower P4 levels. Methodolo-gical problems, however, are associated with thePrior et al. (Prior et al. 1990; De Souza et al. 1997,2003) data set that limit the usefulness of the infor-mation. These limitations include: (i) the inferiormethods used to assess LPD, which included theuse of basal body temperatures and the pooling ofsingle blood samples from the follicular and lutealphases as indicators of ovulatory status; and (ii) theconfusing effect of combining E2-deficient anovu-latory cycles with LPD cycles, since it is likely thatanovulatory cycles have very different E2 produc-tion patterns compared to ovulatory LPD cycles,and thus have the potential to impact bone health.Since then, many researchers have been unable toreproduce the results of Prior et al. (1990) (Lindsay et al. 1978; Winters et al. 1996; De Souza et al. 1997;Waller et al. 1998; Lindsay 1999). Moreover, in post-menopausal women, progestins, whether prescribedalone or in combination with E2, provides neither

an independent nor an additive effect on bone(Lindsay et al. 1978; Lindsay 1999). Simply put, thedata available do not support any significant effect of P4 or LPD on bone. If there is a decrease in bone mass in association with EAMD, such as LPD,and anovulation, it is likely a product of decreasedexposure to E2.

De Souza et al. (1997) have reported that regard-less of significantly lower P4 levels in exercisingwomen with LPD (not combined with women withanovulation), BMD at the lumbar spine and femoralhip were comparable to that observed in ovulat-ory sedentary women, provided the E2 status was adequately maintained during the cycle. In theWomen’s Reproductive Health Study (Waller et al.1998), sedentary women with LPD failed to presentwith reduced BMD as well. One concern is that inthe study by De Souza et al. (1997), significantlylower E2 levels were found in the follicular phase inthe LPD runners. Even in the ovulatory exercisingwomen in that study, E2 levels were lower duringthe early follicular phase (days 2–5). Winters et al.(1996) have also reported reduced E2 levels in theearly follicular phase in trained runners. Import-antly, they also found that the lumbar spine BMDwas lower in their runners with the reductions infollicular phase E2, compared to the active controls.Although this finding was not statistically signific-ant at p = 0.074, it was likely a product of inadequatestatistical power, and the potential physiologicalimportance of the finding should not be dismissed.A limitation of the aforementioned studies is thatthey are cross-sectional and limited by small samplesizes. In the Michigan Bone Health Study, Sowers et al. (1998a, 1998b) found that 31 sedentary womenwho were slightly older than the subjects in the pre-vious studies (mean age of 37 years) and who hadreduced bone mass, also had the lowest E2 levelsduring the luteal phase of two comprehensivelymonitored menstrual cycles.

Thus, it may be necessary to follow exercisingwomen for several years while simultaneously documenting menstrual status and E2 exposure todefinitively answer the important question of long-term effects of LPD and anovulation on BMD. It is likely that women with anovulatory cycles maycertainly experience more serious consequences

Page 285: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 269

due to the greater severity of estrogen depletionassociation with this menstrual anomaly comparedto ovulatory LPD cycles.

Energetics and exercise-associatedmenstrual disturbances: practicalconsiderations

Demographics

With respect to the demographics of EAMD, thehighest prevalence of amenorrhea in athletes isfound in sports that emphasize a low body weightsuch as figure skating, ballet, long-distance run-ning and gymnastics, but more recent studies havedocumented menstrual abnormalities in a widevariety of sports (Erdelyi 1962; Feicht et al. 1978;Dale et al. 1979; Schwartz et al. 1981; Sanborn et al.1982; Loucks & Horvath 1985). Amenorrhea associ-ated with exercise is a typically a diagnosis of exclu-sion; other causes of reproductive disturbances, i.e.pregnancy, androgen excess, gonadal dysgenesis,uterine or premature ovarian failure, or pituitarydysfunction, must be ruled out (Warren 1996). Inphysically active girls who have not experiencedmenarche, primary amenorrhea may ensue despitethese individuals exhibiting normal growth (Warren1996). While no studies have examined the pre-valence of EAMD with increasing age, increasedgynecological maturity may afford some protectionfrom disruption of menstrual cycles (Rogol et al.1992). The majority of studies examining EAMDhave been performed in adolescents or women intheir early 20s. No studies have examined racial orethnic differences in the prevalence or endocrinepresentation of EAMD. Characterizing effects ofrace is important because studies in premenopausaland perimenopasual women show differences insome reproductive hormone concentrations whenCaucasian and African-American women are com-pared (Manson et al. 2001; Reutman et al. 2002), anda high degree of variability when multiple races arestudied (Randolph et al. 2003). Additionally, becausereproductive hormones change in varying directionswith age and increasing body mass index (Randolphet al. 2003), studies examining racial influences mustadjust for these and other (smoking, alcohol use and

physical activity) factors. Another factor pertinentto the assessment of racial differences in EAMD isthat racial and ethnic differences in body image anddieting behavior exist (White et al. 2003). African-American girls may be at a lessened risk for devel-oping eating disorders because they do not displaythe same degree of negative beliefs about body sizeand shape when compared to Caucasian girls (Whiteet al. 2003).

Low energy availability in practical terms

caloric restriction

The evidence for modulation of reproductive func-tion by energy availability in exercising womencomes from short- and long-term prospective stud-ies in humans and animals and from observationaland cross-sectional studies (Bullen et al. 1985;Loucks 1989; Williams et al. 1995; Laughlin & Yen1996; Wade et al. 1996; De Souza et al. 1998a; Loucks et al. 1998; Loucks & Thuma 2003). The suppressionof reproductive function during conditions of lowfuel availability is well-recognized in the animal literature, and is thought to be an adaptive responseto conserve fuel for more vital bodily processes,such as cellular maintenance and locomotion, andprevent the energetically costly investment of gesta-tion and lactation (Wade et al. 1996). A key dif-ference is that a state of low energy availability intoday’s active women may represent a choice toconsciously restrict food intake to a level below thatrequired to match the calories expended throughexercise. Numerous studies have linked EAMD,particularly amenorrhea, with significantly higherscores on psychometric inventories of eating atti-tudes, indicating a conscious restriction of calor-ies associated with a drive for thinness and lowbody fat (Brooks-Gunn et al. 1987; Myerson et al.1991; Wilmore et al. 1992; Laughlin & Yen 1996;Lebenstedt et al. 1999; Warren et al. 1999). Dietary fatis often restricted as well (Laughlin & Yen 1996;Loosli & Ruud 1998). When less severe forms ofmenstrual disturbances are examined, i.e. anovula-tion and LPD, scores on these eating inventories arestill higher than those in subjects who menstruatenormally (Lebenstedt et al. 1999).

Page 286: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

270 chapter 20

physiological cues

While significant associations between weight anddiet concerns and restrictive eating have beenrelated to menstrual disturbances in female athletes,it should not be assumed that every athlete that hasEAMD is consciously restricting her food intake.Many studies describing the endocrine aspects of EAMD included only subjects that had no historyof disordered eating (Loucks et al. 1989, 1992). If anathlete is not consciously restricting her food but is still energy restricted, it is logical to questionwhether the physiological cues to eat are not well-matched to energy needs. Perhaps strenuous exer-cise in some individuals alters the levels of, and/orsensitivity to, physiological cues for hunger or satiety in female athletes, or whether it influencescognitive cues for, or the response to, food intake. In an effort to address a plausible mechanism forinsufficient dietary intake in female athletes, onereport by Hirschberg et al. (1994) found that theresponse of gastrointestinal hormone cholecys-tokinin (CCK), a satiety peptide produced in thegut, was reduced in female long-distance runnerscompared to non-athletes when a 500 kcal (2093 kJ)meal was provided. Increased hunger was alsoreported by the athletes (Hirschberg et al. 1994). Inresponse to acute exercise, Hilton and Loucks (2000)showed that leptin, a signal for satiety, decreaseswith exercise only when energy availability is alsodecreased. When the calories expended as exerciseare compensated for with increased calorie intake,leptin levels did not change significantly. In amen-orrheic athletes, leptin levels are lower than wouldbe expected for their level of body fat (Laughlin & Yen 1997). Recently, the current authors haveinvestigated a link between EAMD and ghrelin, arecently discovered growth hormone secretagoguereleased from the stomach that stimulates hungerand food intake in humans (Date et al. 2000). Ghrelinhas been directly linked to the regulation of energyhomeostasis, and is one of the most powerfulknown orexigens (Nakazato 2001; Wren et al. 2001).Interestingly, one report thus far has linked in-tracerebroventricular ghrelin administration to a suppression of LH pulsatility in ovariectomizedrats, therefore establishing a plausible mechanism

linking low energy availability to a suppression ofmenstrual cyclicity in female athletes (Furuta et al.2003). This is not surprising, as other gut peptides,such as CCK, galanin and peptide YY (PYY) (3–36),have been discovered to exist in the brain and haveneuroendocrine functions (Lopez & Negro-Vilar1990; Challis et al. 2003; Moran & Kinzig 2004). Werecently reported that weight loss resulting from adiet and exercise intervention in young womenleads to an increase in resting levels of ghrelin, andthe change in ghrelin is significantly correlated withthe change in body weight during the intervention(Leidy et al. 2004). In another study we comparedconcentrations of ghrelin between women groupedbased on their exercise and menstrual status. Fast-ing ghrelin levels were assessed in the followinggroups of women: sedentary and ovulating norm-ally; athletes who are ovulating normally; athleteswho are anovulatory or exhibit LPD; and athleteswho are amenorrheic. Fasting ghrelin concentra-tions were found to be significantly elevated in theamenorrheic athletes compared to all other groups(De Souza et al. 2004). Because the amenorrheic ath-letes in the study exhibited other endocrine signs of energy deficiencyai.e. low levels of triiodothyro-nine, insulin, leptin and other alterations in meta-bolics hormones (Fig. 20.3)abut had been weightstable, it appears that physiological cues to stimu-late food intake are responding appropriately to astate of low energy availability, whereas other cues,perhaps cognitive in nature, are preventing a returnto energy homeostasis. This would agree with thefindings of elevated concentrations of ghrelin inanorexics (Otto et al. 2001). With respect to how lowenergy availability develops in an active woman,much still needs to be learned in order to identifythe predisposing factors that predict which athleteswill consciously restrict their calories in addition totheir training, and which athletes may simply not beable to, or physiologically stimulated to, increasetheir food intake to meet their energy needs.

prediction of exercise-associated

menstrual disturbances based on

energy intake and expenditure

Despite advances in our understanding of the

Page 287: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 271

Eumetabolic state? Intermittent/transient

hypometabolic state Hypometabolic state

Ovulatory menstrual cycles

– Ghrelin

Total T3

Leptin

– Insulin

– Growth hormone

– IGF-I/IGFBP-1

Cortisol

– Glucose

Luteal phase deficientmenstrual cycles

Metabolic hormones and substrates relative to sedentary women

Amenorrheic cycles

– Ghrelin

Total T3

Leptin

Insulin

Growth hormone

– IGF-I/IGFBP-1

Cortisol

– Glucose

Ghrelin

Total T3

Leptin

Insulin

Growth hormone

IGF-I/IGFBP-1

Cortisol

Glucose

LH pulsatility

– FSH

Estradiol

Progesterone

Reproductive hormones relative to sedentary women

LH pulsatility

FSH

Estradiol

Progesterone

LH pulsatility

FSH

Estradiol

Progesterone

Fig. 20.3 The metabolic and reproductive hormone perturbations that have been identified to date and associated withexercise training and menstrual status, including eumenorrheic ovulatory cycles, luteal phase defect (LPD) cycles andamenorrhea. All values are depicted by arrows signifying the magnitude of the alteration reported. The proposedrelationship to menstrual status is also shown. The repeated transitions from ovulatory cycles and LPD cycles are shown.Although not documented in the literature, it is likely that during recovery from amenorrhea, an individual is likely toexperience LPD cycles. FSH, follicle-stimulating hormone; LH, luteinizing hormone; IGF-I, insulin-like growth factor I;IGFBP-1, insulin-like growth factor binding protein 1; T3 triiodothyronine. (Modified and reprinted with permission fromDe Souza 2003.)

Page 288: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

272 chapter 20

causal role that low energy availability plays in theetiology of EAMD, we have not yet developed practical guidelines regarding the optimal com-bination(s) of dietary intake and exercise energyexpenditure to maintain ovulation in active women.Although it is believed that female athletes may nothave to reduce their training to avoid EAMD, avoiding disruptions in menstrual function wouldconceivably necessitate the quantification of foodintake and energy expenditure in order to determinehow much food is required to match the level ofcalories expended. Because efforts to optimize per-formance may predispose an athlete to take in theminimum amount of calories required to preventfatigue but maintain ovulation, it would be helpfulto know ‘how low you could go’ without comprom-ising reproductive function. Many (Drinkwater et al.1984; Marcus 1985; Kaiserauer et al. 1989) but not all (Loucks 1989; Wilmore et al. 1992) studies of self-reported calorie intake in amenorrheic athletesreport that calorie intake is lower than expected for the level of estimated energy expenditure, andlower when compared to eumenorrheic athleteswith similar training regimens. It is difficult totranslate the findings of these studies into advice forpractitioners, because there is a tendency for mostsubjects to under-report food intake. Regarding thesuppression of reproductive function associatedwith calorie restriction, neither the magnitude of the reduction in the calories consumed, nor theabsolute amount of calories consumed appears to be what is sensed by gonadotropin-releasing hor-mone (GnRH) neurons. Support for this comes fromLoucks’ studies where short-term energy deficitsare created by varying combinations of food intakeand exercise and the resulting suppression of LHpulsatility is dependent on the magnitude of theenergy deficit, regardless of if it is created throughexercise or diet, or a combination (Loucks & Thuma2003). Additionally, in studies in exercising mon-keys, food intake remained constant as amenorrheadeveloped due solely to increases in the energy cost of daily exercise (Williams et al. 2001a). Thisdependence of EAMD on energy availability per se,and not a particular level of calorie intake or amountof exercise, presents a problem with regard to thedetection and prevention of EAMD. It means that a

wide range exists in the volume of exercise and ordaily calorie intake that is associated with EAMD,and prevents either from being used as a bench-mark. Perhaps the most progress in actually quantifying changes in energy intake and energyexpenditure associated with changes in the repro-ductive axis in exercising women has been made byLoucks et al. (Loucks & Thuma 2003). A threshold ofenergy availability between 20 and 30 kcal·kg–1 (84and 126 kJ·kg−1) lean body mass was identified suchthat no effect on the reproductive axis was observeduntil this threshold was achieved. When energyavailability of exercising subjects was held between20 and 30 kcal·kg–1 (84 and 126 kJ·kg−1) lean bodymass, LH pulsatility, a proxy indicator of the GnRHpulse generator, was decreased. Importantly, becausethe initial energy availability of the subjects was 45 kcal·kg–1 (188 kJ·kg−1) lean body mass, these dataalso show that a decrease of energy availability by33% had no effect on LH pulsatility, suggesting thatthe female reproductive axis can withstand substan-tial reductions in energy availability before thereproductive axis responds. Additionally, the datasuggest that no change in LH pulsatility occurredeven though exercise calories were as high as 840 kcal (3516 kJ), which might represent an 8-mile(13-km) run. However, whether a similar thresholdto that identified by Loucks’ short-term studiesholds true for the effects of prolonged training onovulation and menstrual cyclicity remains to bedemonstrated. Further, whether the concept of a‘threshold’ holds true for the development of lesssevere menstrual disturbances, such as LPD, isunknown. Ongoing studies aimed at inducing anquantifiable energy deficit with exercise combinedwith calorie restriction and perturbing the men-strual cycle are underway and may, in the future,provide practical guidelines for athletes and activewomen.

prediction of exercise-associated

menstrual disturbances based on

body weight and body fat

Predicting EAMD would be much easier if changesin reproductive function occurred at a predictablelevel of body weight and or body fat, as originally

Page 289: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 273

proposed by Frisch and Revelle (1971). Bullen et al.(1985), in a prospective exercise training study overtwo menstrual cycles, showed that the incidence of EAMD increases with weight loss that aver-aged –0.45 kcal·wk–1 (−1.88 kJ·wk−1) and –4.0 + 0.3 kgover the course of the study. Whether this rate andmagnitude of weight loss represent potential guide-lines for women just beginning an exercise programis questionable, since no individual correlationswere reported between the magnitude of weightloss and the severity of menstrual disturbances, andmenstrual cyclicity was disrupted in the group thatmaintained body weight. Other studies have shownthat EAMD exist in women who represent a range of body weights and levels of percentage body fat(Sanborn et al. 1987), and the induction of amenor-rhea can occur with no change in body weight(Williams et al. 2001a). Further, changes in repro-ductive hormone secretion resulting from short-term changes in nutritional intake occur withoutsignificant weight loss or gain (Bronson 1986;Cameron & Nosbisch 1991). Lastly, the effect ofweight loss or the loss of body fat on reproductivefunction may or may not be dependent on initialbody fat stores (Alvero et al. 1998), thereby imposingan additional interaction to consider. Therefore,advising female athletes not to lose a certain amountof weight to prevent EAMD, or to gain a certainamount of weight to restore menstrual cyclicity, isnot reliable. Guidelines regarding the prevention of EAMD based on body weight are further com-plicated because of the large individual variabilityin the amount of weight lost in response to specificenergy deficits (Ravussin et al. 2001).

reversing exercise-associated

menstrual disturbances by increasing

energy availability

To date, the only non-medical advice provided forprevention and treatment of amenorrhea is that athletes reduce their training and increase theircaloric intake. No specific dietary guidelines areavailable, and the rationale for reducing trainingmay be an overly conservative approach, since exercise per se does not appear to play a role in exercise-associated amenorrhea. Quantification of

the magnitude of change in energy availability associated with the induction of EAMD may bemore difficult to achieve than quantification of themagnitude of change in energy availability requiredto restore menstrual cyclicity. One reason is thatresearchers have been more successful at document-ing the reversal of amenorrhea (Drinkwater et al.1986; Dueck et al. 1996; Kopp-Woodroffe et al. 1999;Perkins et al. 2001; Zeni-Hoch et al. 2003) in exercis-ing women than inducing amenorrhea with a dietand/or exercise intervention (Williams et al. 2001a).In exercising monkeys who had developed amenor-rhea, Williams et al. (2001b) showed that, unlike theinduction of amenorrhea, the reversal (as noted byovulation) exhibited a linear dose–response relation-ship with increased energy availability, such thatthe two monkeys that consumed 163% and 181% oftheir baseline intake recovered in 16 and 12 days,respectively, and two monkeys that consumed138% and 141% recovered in 57 and 50 days, respect-ively. The time in days required for recovery wasinversely related to the amount of extra calories consumed by each monkey (r = –0.97; P < 0.02). Incontrast to the lack of change in body weight duringthe induction of EAMD, body weight increasedsignificantly with the restoration of ovulation andsubsequent menses. There was a significant correla-tion between body weight during the amenorrheicperiod and the time to recovery, i.e. the monkeysthat weighed less recovered more quickly than theheavier monkeys, but there was no correlationbetween the magnitude of change in weight and the rapidity of the restoration. Overall, body weightincreased 3–11%. Notably, the time to recovery forthese monkeys was much shorter than the time ittook to become amenorrheic.

Although the reversal of exercise-associated ame-norrhea has been documented by several studies inhumans, there have only been two small studiespublished on the reversal of amenorrhea attemptedby a supervised intervention that modified diet andexercise (Dueck et al. 1996; Kopp-Woodroffe et al.1999). Dueck et al. (1996) were the first to publish theresults of a reversal of amenorrhea intervention inan athlete consisting of a 15-week diet and exerciseintervention. The intervention consisted of increas-ing daily energy intake by 360 kcal (1507 kJ) and

Page 290: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

274 chapter 20

decreasing exercise training by 1 day per week. Theincrease in energy intake was accomplished with aliquid supplement. At the end of the 15-week inter-vention, the subject remained amenorrheic, despitea 2.7 kg increase in body weight and increased bodyfat from 8% to 14%, likely a product of improvedand positive energy balance from –155 kcal·day–1

(−648 kJ·day−1) at baseline to +683 kcal·day–1 (+2855kJ·day−1) accomplished by the end of the 15-weekintervention. The intervention also resulted in a 148%increase in serum LH levels. Interestingly, menstru-ation did resume in this amenorrheic athlete after an additional self-initiated 3 months of additionalcompliance to the protocol. A second study (Kopp-Woodroffe et al. 1999) attempted the reversal ofamenorrhea with a 20-week diet and intervention infour amenorrheic athletes. The women were againprovided with a 360 kcal·day–1 (1507 kJ·day−1) liquidsupplement and asked to restrict exercise training for1 day per week. Three of the four subjects resumednormal menstrual function during or shortly follow-ing the intervention period, again likely a result of theimproved energy balance of +164–292 kcal·day–1

(+686–1222 kJ·day−1). The fourth subject withdrewfrom the intervention to begin hormone therapy forpoor bone density. As reported in the study results,time to menstruation subsequent to ovulation inthese three subjects ranged from 13 to 24 weeks.Using data reported for calorie intake and expend-iture, the estimated changes in energy balanceranged from +8% to +16%.

The data from the previous studies provide themost useful information to date on how to achievethe restoration of menstrual cyclicity in amenorrheicathletes. An important consideration in attempting

to reverse amenorrhea with an ‘energy intervention’is that the alteration in diet, exercise, or both, be tailored to the individual’s own preference. Forsome individuals, a reduction in training may not beas well-tolerated psychologically as an increase incalories, but for others a combination or only achange in training may be preferred. Careful mon-itoring of both exercise and diet is important sincethere may be a tendency for an athlete to unknow-ingly compensate for an increase in food intake with an increase in training frequency or duration(Kopp-Woodroffe et al. 1999). In the latter study,two out of four subjects could not completelyadhere to the reduction in training called for by theintervention.

Summary and conclusions

EAMD can be associated with significant clinicaloutcomes, and thus continued efforts aimed atidentification and prevention is necessary. Bone loss and endothelial dysfunction has been linked to hypoestrogenism secondary to amenorrhea infemale athletes. The extent to which less severeEAMD are associated with changes in bone and cardiovascular function remain unclear, but is a fertile area for future research. The cause of EAMDis not completely understood but a causal relation-ship has been established between low energy availability and the induction of menstrual dis-turbances. Recovery of normal menstrual functionappears to depend directly on the magnitude ofchange in energy availability, accomplished throughan increase in food intake that may or may not becombined with decreased exercise training.

References

Alvero, R., Kimzey, L., Sebring, N. et al. (1998) Effects of fasting onneuroendocrine function and follicledevelopment in lean women. Journal ofClinical Endocrinology and Metabolism 83,76–80.

Armour, K.E. & Ralston, S.H. (1998)Estrogen upregulates endothelialconstitutive nitric oxide synthaseexpression in human osteoblast-likecells. Endocrinology 139, 799–802.

Arora, S., Veves, A., Caballaro, A.E.,Smakowski, P.A. & LoGerfo, F.W. (1998)Estrogen improves endothelial function.Journal of Vascular Surgery 27(6),1141–1146.

Ayres, S., Baer, J., Subbiah, M.T. & Ravi, H.(1998). Exercised-induced increase inlipid peroxidation parameters inamenorrheic female athletes. Fertility andSterility 69(1), 73–77.

Baker, E.R., Mathur, R.S., Kirk, R.F. &

Williamson, H.O. (1981) Female runnersand secondary amenorrhea: correlationwith age, parity, mileage, and plasmahormonal and sex-hormone bindingglobulin concentrations. Fertility andSterility 36, 183–187.

Balash, J. & Vanrell, J.A. (1987) Corpusluteum insufficiency and infertility: a matter of controversy. HumanReproduction 2, 557–567.

Beitins, I.Z., McArthur, J.W., Turnbull,

Page 291: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 275

B.A., Skrinar, G.S. & Bullen, B.A. (1991)Exercise induces two types of humanluteal dysfunction: conformation byurinary free progesterone. Journal ofClinical Endocrinology and Metabolism 72,1350–1358.

Blacker, C.M., Ginsburg, K.A., Leach, R.E.,Randolph, J. & Moghissi, K.S. (1997)Unexplained infertility: evaluation ofthe luteal phase; results of the NationalCenter for Infertility Research atMichigan. Fertility and Sterility 67(3),437–442.

Blumel, J.E., Castelo-Branco, C., Leal, T. et al. (2003) Effects of transdermalestrogens on endothelial function in postmenopausal women withcoronary disease. Climacteric 6(1), 38–44.

Bronson, F.H. (1986) Food-restricted,preubertal, female rats: rapid recoveryof luteinizing hormone pulsing withexcess food, and full of pubetaldevelopment with gonadotropin-releasing hormone. Endocrinology 118,2483–2487.

Broocks, A., Pirke, K.M., Schweiger, U. et al. (1990) Cyclic ovarian function inrecreational athletes. Journal of AppliedPhysiology 68(5), 2083–2086.

Brooks-Gunn, J., Warren, M.P. &Hamilton, L.H. (1987) The relation ofeating problems and amenorrhea inballet dancers. Medicine and Science inSports and Exercise 19(1), 41–44.

Bullen, B.A., Skrinar, G.S., Beitins, I.Z. et al.(1985) Induction of menstrual disordersby strenuous exercise in untrainedwomen. New England Journal of Medicine312, 1349–1353.

Cameron, J.L. & Nosbisch, C. (1991)Suppression of luteinizing hormone andtestosterone secretion during short termfood restriction in the adult male rhesusmonkey (Macaca mulatta). Endocrinology128, 1532–1540.

Cann, C.E., Mortin, M.C., Genant, H.K. & Jaffe, R.B. (1984) Decreased spinemineral content in amenorrheic women.Journal of the American Medical Association251, 626–629.

Carbon, R., Sambrook, P.N., Deakin, V. et al. (1990) Bone density of elite femaleathletes with stress fractures. MedicalJournal of Australia 153, 373–376.

Celermajer, D.S. Endothelial dysfunction:does it matter? Is it reversible? (1997)Journal of the American College ofCardiology 30(2), 325–333.

Celermajer, D.S., Sorensen, K.E., Gooch,V.M. et al. (1992) Non-invasive detectionof endothelial dysfunction in children

Endocrinology and Metabolism 83,4220–4232.

De Souza, M.J., Miller B.E., Luciano, A.A.et al. (1998b) Letter to the editor.Ovulation and spine bone mineraldensityaauthor’s response. Journal ofClinical Endocrinology and Metabolism83(10), 3757–3758.

De Souza, M.J., vanHeest, J.L., Demers, L.& Lasley, B.L. (2003) Luteal phasedeficiency in recreational runners:evidence for a hypometabolic state.Journal of Clinical Endocrinology andMetabolism 88(1), 337–346.

De Souza, M.J., McConnell, H.J.,O’Donnell, E., Lasley, B.L. & Williams,N.I. (2004) Fasting ghrelin levels inphysically active women: relationshipwith menstrual disturbances andmetabolic status. Journal of ClinicalEndocrinology and Metabolism 10(5),433–448.

Drew, F.L. (1961) The epidemiology ofsecondary amenorrhea. Journal ofChronic Disease 14, 396–401.

Drinkwater, B.L, Nilson, K., Chesnut, C.H.et al. (1984) Bone mineral content ofamenorrheic and eumenorrheic athletes.New England Journal of Medicine 311,277–281.

Drinkwater, B.L., Nilson, K., Ott, S. &Chestnut, C.H., 3rd. (1986) Bone mineraldensity after resumption of menses inamenorrheic athletes. Journal of theAmerican Medical Association 256,380–382.

Drinkwater, B.L., Bruemner, B. &Chestnut, C.H., 3rd. (1990) Menstrualhistory as a determinant of current bonedensity in young athletes. Journal of theAmerican Medical Association 263(4),545–548.

Dueck, C.A., Matt, K.S., Manore, M.M. &Skinner, J.S. (1996) Treatment of athleticamenorrhea with a diet and trainingintervention program. InternationalJournal of Sports Nutrition 6, 24–40.

Dugowson, C.E., Drinkwater, B.L. & Clark,J.M. (1991) Nontraumatic femur fracturein an oligomenorrheic athlete. Medicineand Science in Sports and Exercise 23,1323–1325.

Ellison, P.T. & Lager, C. (1986) Moderaterecreational running is associated withlowered salivary progesterone profilesin women. American Journal of Obstetricsand Gynecology 154, 1000–1003.

Erdelyi, G.J. (1962) Gynecological surveyof female athletes. Journal of SportMedicine and Physical Fitness 2, 174–197.

Fagan, K.M. (1998) Pharmacologic

and adults at risk of atherosclerosis.Lancet 340(8828), 1111–1115.

Celermajer, D.S., Sorensen, K.E.,Spiegelhalter, D.J. et al. (1994) Aging isassociated with endothelial dysfunctionin healthy men years before the age-related decline in women. Journal of theAmerican College of Cardiology 24(2),471–476.

Challis, B.G., Pinnock, S.B., Coll, A.P. et al.(2003) Acute effects of PYY3–36 on foodintake and hypothalamic neuropeptideexpression in the mouse. Biochemical andBiophysical Research Communications311(4), 915–919.

Cheung, C.C., Thornton, J.E., Kuiper, J.L. et al. (1997) Leptin is a metabolic gate forthe onset of puberty in the female rat.Endocrinology 138, 855–858.

Cobb, K.L., Bachrach, L.K., Greendale, G. et al. (2003) Disordered eating, menstrualirregularity, and bone mineral density infemale runners. Medicine and Science inSports and Exercise 35(5), 711–719.

Csermely, T., Halvax, L., Schmidt, E. et al. (2002) Occurrence of osteopeniaamong adolescent girls witholigo/amenorrhea. Gynecology andEndocrinology 2, 99–105.

Cumming, D.C. (1996) Exercise-associatedamenorrhea, low bone density, andestrogen replacement therapy. Archivesof Internal Medicine 156, 2193–2195.

Dale, E., Gerlack, D.H. & Wilhite, A.L.(1979) Menstrual dysfunction in distancerunners. Obstetrics and Gynecology 54,47–53.

Date, Y., Kojima, M., Hosoda, H. et al. (2000) Ghrelin, a novel growthhormone-releasing acylated peptide, issynthesized in a distinct endocrine celltype in the gastrointestinal tracts of rats and humans. Endocrinology 141,4255–4261.

De Souza, M.J. (2003) Menstrualdisturbances in athletes: a focus on lutealphase defects. Medicine and Science inSports and Exercise 35(9), 1553–1563.

De Souza, M.J., Miller, B.E., Sequenzia,L.C. et al. (1997) Bone health is notaffected by luteal phase abnormalitiesand decreased ovarian progesteroneproduction in female runners. Journal ofClinical Endocrinology and Metabolism 82,2867–2876.

De Souza, M.J., Miller, B.E., Loucks, A.B. et al. (1998a) High frequency of lutealphase deficiency and anovulation inrecreational women runners: bluntedelevation in follicle-stimulatinghormone observed during luteal-follicular transition. Journal of Clinical

Page 292: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

276 chapter 20

management of athletic amenorrhea.Clinics in Sports Medicine 17, 327–331.

Feicht, C.B., Johnson, T.S., Martin, B.J. et al. (1978) Secondary amenorrhea in athletes. Lancet 2, 1145–1146.

Frisch, R.E. & Revelle, R. (1971) Height andweight at menarche and a hypothesis of menarche. Archives of Disease inChildhood 46, 695–701.

Frisch, R.E., Wyshak, G. & Vincent, L.(1980) Delayed menarche andamenorrhea in ballet dancers. NewEngland Journal of Medicine 303(1), 17–19.

Furuta, M., Funabashi, T., Kimura, F.(2003) Intro cerebroventricularadministration of ghrelin rapidlysuppresses pulsatile luteinizinghormone secretion in ovariectomizedrats. Biochemical and Biophysical ResearchCommunications 288(4), 780–785.

Gremion, G., Rizzoli, R., Slosman, D.,Theintz, G. & Bonjour, J.P. (2001) Oligo-amenorrheic long-distance runners maylose more bone in spine than femur.Medicine and Science in Sports and Exercise33(1), 15–21.

Hamilton-Fairly, D. & Taylor, A. (2003)Anovulation. BMJ (Clinical Research Ed.)327(7414), 546–549.

Hilton, L.K. & Loucks, A.B. (2000) Lowenergy availability, not the stress ofexercise, suppresses the diurnal rhythmof leptin in healthy young women.American Journal of Physiology.Endocrinology and Metabolism 278(1),E43–E49.

Hirschberg, A.L., Lindholm, C., Carlstrom,K. & Von Schoultz, B. (1994) Reducedserum cholecystokinin response to foodintake in female athletes. Metabolism43(2), 217–222.

Jones, G.S. (1976) The luteal phase defect.Fertility and Sterility 27, 351–356.

Jones, G.S. & Madigal-Castro, V. (1970)Hormonal findings in association withabnormal corpus luteum function in thehuman: the lueal phase defect. Fertilityand Sterility 21, 1–13.

Jonnavithula, S., Warren, M.P., Fox, R.P. & Lazaro, M. (1993) Bone density iscompromised in amenorrheic womendespite return of menses: a 2-year study.Obstetrics and Gynecology 81(5), 669–674.

Kaiserauer, S., Snyder, A.C., Sleeper, M. & Zierath, J. (1989) Nutritional,physiological, and menstrual status ofdistance runners. Medicine and Science inSports and Exercise 21(2), 120–125.

Kanis, J.A. (2002) Diagnosis ofosteoporosis and assessment of fracturerisk. Lancet 359, 1929–1936.

diets in female athletes: a red flag.Physician and Sports Medicine 26(11),45–48.

Lopez, F.J. & Negro-Vilar, A. (1990)Galanin stimulates luteinizing hormone-releasing hormone secretion fromarcuate nucleus-median eminencefragments in vitro: involvement of an α-adrenergic mechanism. Endocrinology127(5), 2431–2436.

Loucks, A.B. (1989) Alterations in thehypothalamic–pituitary–ovarian andthe hypothalamic–pituitary–adrenalaxes in athletic women. Journal of ClinicalEndocrinology and Metabolism 68(2),402–411.

Loucks, A.B. & Horvath, S.M. (1985)Athletic amenorrhea: a review. Medicineand Science in Sports and Exercise 17(1),56–72.

Loucks, A.B. & Thuma, J.R. (2003)Luteinizing hormone pulsatility isdisrupted at a threshold of energyavailability in regularly menstruatingwomen. Journal of Clinical Endocrinologyand Metabolism 88, 297–311.

Loucks, A.B., Laughlin, G.A., Mortola, J.F.et al. (1992) Hypothalamic–pituitary–thyroidal function in eumenorrheic andamenorrheic athletes. Journal of ClinicalEndocrinology and Metabolism 75,514–518.

Loucks, A.B., Verdun, M. & Heath, E.M.(1998) Low energy availability, not stressof exercise, alters LH pulsatility inexercising women. Journal of AppliedPhysiology 84, 37–46.

Luscher, T.F. & Barton, M. (1997) Biologyof the endothelium. Clinical Cardiology20(11) (suppl. 2), II-3–II-10.

McNeely, M.J. & Soules, M.R. (1988) Thediagnosis of luteal phase deficiency: acritical review. Fertility and Sterility 50,1–15.

Malcolm, C.E. & Cumming, D.C. (2003)Does anovulation exist in eumenorrheicwomen? Obstetrics and Gynecology102(2), 317–318.

Malina, R.M. (1994) Physical growth andbiological maturation of young athletes.Exercise and Sport Science Reviews 22,389–433.

Manson, J.M., Sammel, M.D., Freeman,E.W. & Grisso, J.A. (2001) Racialdifferences in sex hormone levels inwomen approaching the transition tomenopause. Fertility and Sterility 75(2),297–304.

Marcus, R., Cann, C., Madvig, P. et al.(1985) Menstrual function and bonemass in elite women distance runners.Annals of Internal Medicine 102, 158–163.

Keen, A.D. & Drinkwater, B.L. (1997)Irreversible bone loss in formeramenorrheic athletes. OsteoporosisInternational 7, 311–315.

Khan, K.M, Liu-Ambrose, T., Sran, M.M. et al. (2002) New criteria for the femaleathlete triad syndrome? British Journal of Sports Medicine 36(1), 10–13.

Kopp-Woodroffe, S.A., Manore, M.M.,Dueck, C.A., Skinner, J.S. & Matt, K.S.(1999) Energy and nutrient status ofamenorrheic athletes participating in adiet and exercise training interventionprogram. International Journal of SportsNutrition 9, 70–88.

Korpelainen, R., Orava, S., Karpakka, J.,Siira, P.& Hulkko, A. (2001) Risk factorsfor recurrent stress fractures in athletes.American Journal of Sports Medicine 29(3),304–310.

Lauder, T.O., Williams, M.V., Campbell,C.S. et al. (1999) The female athlete triad:prevalence in military women. MilitaryMedicine 164, 630–635.

Laughlin, G.A. & Yen, S.S.C. (1996)Nutritional and endocrine-metabolicaberrations in amenorrheic athletes.Journal of Clinical Endocrinology andMetabolism 81, 4301–4309.

Laughlin, G.A. & Yen, S.S.C. (1997)Hypoleptinemia in women athletes:absence of a diurnal rhythm withamenorrhea. Journal of ClinicalEndocrinology and Metabolism 82,318–321.

Lebenstedt, M., Platte, P. & Pirke, K. (1999) Reduced resting metabolic rate in athletes with menstrual disorders.Medicine and Science in Sports and Exercise31(9), 1250–1256.

Leidy, H.J., Gardner, J.K., Frye, B.R. et al.(2004) Circulating ghrelin is sensitive tochanges in body weight during a dietand exercise program in normal weightyoung women. Journal of ClinicalEndocrinology and Metabolism 89(6),2659–2664.

Lindsay, R. (1999) The lack of effect ofprogesterone on bone. Journal of SportsMedicine 44(2), 215–220.

Lindsay, R., Hart, D.M., Purdie, D. et al.(1978) Comparative effects of oestrogenand a progesterone on bone loss inpostmenopausal women. ClinicalSciences and Molecular Medicine 54,193–195.

Lipson, S.F. & Ellison, P.T. (1996)Comparison of salivary steroid profilesin naturally occurring conception andnon-conception cycles. HumanReproduction 11(10), 2090–2096.

Loosli, A.R. & Ruud, J.S. (1998) Meatless

Page 293: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

energy balance and the menstrual cycle 277

Matkovic, V., Jelic, T., Wardlow, G.M. et al.(1994) Timing of peak bone mass inCaucasian females and its implicationfor the prevention of osteoporosis.Inference from a cross-sectional model.Journal of Clinical Investigation 93,799–808.

Mendelsohn, M.E. & Karas, R.H. (1999)The protective effects of estrogen on thecardiovascular system. New EnglandJournal of Medicine 340(23), 1801–1811.

Moran, T.H. & Kinzig, K.P. (2004)Gastrointestinal satiety signals II.Cholecystokinin. American Journal ofGastrointestinal Liver Physiology 286(2),G183–G188.

Myerson, M., Gutin, B., Warren, M.P. et al. (1991) Resting metabolic rate andenergy balance in amenorrheic andeumenorrheic runners. Medicine andScience in Sports and Exercise 23, 15–22.

Nakazato, M., Murakami, N., Date, Y. et al.(2001) A role of ghrelin in the centralregulation of feeding. Nature 409,194–198.

O’Donnell, E., Goodman, J., Witt, J. et al.(2004) Hypoestrogenism in amenorrheicathletes modulates cardiovascularfunction. Proceedings from theAmerican College of Sports MedicineAnnual Meeting. Medicine and Science inSports and Exercise 36(5), suppl., 533.

Ohmichi, M., Kanda, Y., Hisamoto, K. et al. (2003) Rapid changes of flow-mediated dilatation after surgicalmenopause. Maturitas 44(2), 125–131.

Otis, C.L., Drinkwater, B.L., Johnson, M.,Loucks, A.B. & Wilmore, J.H. (1997)ACSM Position Stand on the femaleathlete triad. Medicine and Science inSports and Exercise 29(5), i–ix.

Ott, S.M. (1990) Attainment of peak bonemass. Journal of Clinical Endocrinology and Metabolism 71, 1082A–1082C.

Otto, B., Cuntz U., Fruehauf, E. et al. (2001)Weight gain decreases elevated plasmaghrelin concentrations of patients withanorexia nervosa. European Journal ofEndocrinology 145, R5–R9.

Petterson, F., Fires, H. & Nillius, S.J. (1973) Epidemiology of secondaryamenorrhea: prevalence and incidencerates. American Journal of Obstetrics andGynecology 117, 80–86.

Pettersson, U., Stalnacke, B., Ahlenius, G.et al. (1999) Low bone mass density atmultiple skeletal sites, including theappendicular skeleton in amenorrheicrunners. Calcified Tissue International 64,117–25.

Perkins, R.B., Hall, J.E., Martin, K.A. (2001)Aetiology, previous menstrual function

& Gatz, M. (1979) The relationshipbetween long distance running, plasmaprogesterone and luteal phase length.Fertility and Sterility 31, 130–133.

Sherman, B.M. & Korenman, S.G. (1974)Measurement of plasma LH, FSH,estradiol, and P4 in disorders of thehuman menstrual cycle: the short lutealphase. Journal of Clinical Endocrinologyand Metabolism 38, 89–93.

Simoncini, T., Varone, G., Fornari, L. et al. (2002) Genomic and nongenomicmechanisms of nitric oxide synthesisinduction in human endothelial cells bya fourth-generation selective estrogenreceptor modulator. Endocrinology143(6), 2052–2061.

Soules, M.R. (1988) Luteal-phasedeficiency: The most commonabnormality of the menstrual cycle? In: The Menstrual Cycle and Its Disorders(Pirke, K.M., Wuttke, W. & Schweiger,U., eds.). Springer-Verlag, New York:97–109.

Sowers, M., Crutchfield, M., Bandekar, R.et al. (1998a) Bone mineral density andits change in pre- and perimenopausalwhite women: the Michigan bone healthstudy. Journal of Bone and MineralResearch 13, 1134–1140.

Sowers, M., Randolph, J.F., Crutchfield, M.et al. (1998b) Urinary ovarian andgonadotropin hormone levels inpremenopausal women with low bonemass. Journal of Bone and Mineral Research13, 1191–1202.

Steinberg, D.S. (1987) Lipoproteins and the pathogenesis of atherosclerosis.Circulation 76, 508–514.

Strott, C.A., Cargille, C.M., Ross, G.T. &Lipsett, M.B. (1970) The short lutealphase. Journal of Clinical Endocrinologyand Metabolism 30, 246–251.

Tomten, S.E., Falch, J.A., Birkeland, K.I.,Hemmersbach, P. & Hostmark, A.T.(1998) Bone mineral density andmenstrual irregularities. A comparativestudy on cortical and trabecular bonestructures in runners with allegednormal eating behavior. InternationalJournal of Sports Medicine 19(2), 92–97.

Veldhuis, J.D., Evans, W.S., Demers, L.M.et al. (1985) Altered neuroendocrineregulation of gonadotropin secretion in women distance runners. Journal ofClinical Endocrinology and Metabolism 61,557–563.

Wade, G.N., Schneider, J.E. & Hui-Yun, L.(1996) Control of fertility by metaboliccues. American Journal of Physiology.Endocrinology and Metabolism 33(270),E1–E9.

and patterns of neuro-endocrinedisturbance as prognostic indicators inhypothalamic amenorrhea. HumanReproduction 16(10), 2198–2205.

Prior, J.C., Vigna, Y.M., Schechter, M.T. &Burgess, A.E. (1990) Spinal bone loss andovulatory disturbances. New EnglandJournal of Medicine 323, 1221–1227.

Randolph, J.F., Sowers, M., Gold, E.B. et al.(2003) Reproductive hormones in earlymenopausal transition: relationship toethnicity, body size, and menopausalstatus. Journal of Clinical Endocrinologyand Metabolism 88, 1516–1522.

Ravussin, E., Tschop, M., Morales, S.,Bouchard, C. & Heiman, ML (2001)Plasma ghrelin concentration andenergy balance: overfeeding andnegative energy balance studies intwins. Journal of Clinical Endocrinologyand Metabolism 86, 4547–4551.

Reutman, S.R., LeMasters, G.K., Kesner,J.S. et al. (2002) Urinary reproductivehormone level difference betweenAfrican American and Caucasianwomen of reproductive age. Fertility andSterility 78(2), 383–391.

Rogol, A.D., Weltman, A., Weltman, J.Y. et al. (1992) Durability of thereproductive axis in eumenorrheicwomen during 1 year of endurancetraining. Journal of Applied Physiology72(4), 1571–1580.

Rutherford, O.M. (1993) Spine and total body bone mineral density inamenorrheic endurance athletes. Journalof Applied Physiology 74, 2904–8.

Sanborn, C.F., Martin, B.J. & Wagner,W.W., Jr. (1982) Is athletic amenorrheaspecific to runners? American Journal ofGynecology 143, 859–863.

Sanborn, C.F, Albrecht, B.H. & Wagner,W.W. (1987) Athletic amenorrhea: lackof association with body fat. Medicineand Science in Sports and Exercise 19(3),207–212.

Sanborn, C.F., Horea, M., Siemers, B. &Dieringer, K. (2000) Disordered eatingand the female athlete triad. Clinics inSport Medicine 19(2), 199–213.

Schachinger, V., Britten, M.B. & Zeiher,A.M. (2000) Prognostic impact ofcoronary vasodilator dysfunction on adverse long-term outcome ofcoronary heart disease. Circulation 101,1899–1906.

Schwartz, B., Cumming, D.C., Riordan, E.et al. (1981) Exercise-associatedamenorrhea: a distinct entity? AmericanJournal of Obstetrics and Gynecology 141,662–670.

Shangold, M.M., Freeman, R., Thysen, B.

Page 294: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

278 chapter 20

Waller, K., Reim, J., Fenster, L. et al. (1996)Bone mass and subtle abnormalities inovulatory function in healthy women.Journal of Clinical Endocrinology andMetabolism 81, 663–668.

Waller, K., Swan, S.H., Windham, G.C. et al. (1998) Use of urine biomarkers toevaluate menstrual function in healthypremenopausal women. AmericanJournal of Epidemiology 147(11),1071–1080.

Warren, M.P. (1980) The effects of exercise on pubertal progression andreproductive function in girls. Journal ofClinical Endocrinology and Metabolism 51,1150–1157.

Warren, M.P. (1996) Evaluation ofsecondary amenorrhea. Journal of ClinicalEndocrinology and Metabolism 81(2),437–442.

Warren, M.P. Voussoughian, F., Geer, E.B.et al. (1999) Functional hypothalamicamenorrhea: hypoleptinemia anddisordered eating. Journal of ClinicalEndocrinology and Metabolism 84,873–877.

White, M.A., Kohlmaier, J.R., Varnado-Sullivan, P. & Williamson, D.A. (2003)Racial/ethnic differences in weightconcern: protective and risk factors forthe development of eating disorders and obesity among adolescent females.Eating and Weight Disorders 8(1), 20–25.

Williams, N.I. (1998) Low energyavailability and reproductivedisturbances: a review of clinical andhormonal effects. Journal of DanceMedicine and Science 2(1), 19–31.

conservation in amenorrheic comparedwith eumenorrheic distance runners?Journal of Applied Physiology 72(1), 15–22.

Winters, K.M., Adams, W.C., Meredith,C.N., Van Loan, M.D. & Lasley, B.L.(1996) Bone density and cyclic ovarianfunction in runners and active controls.Medicine and Science in Sports and Exercise28(7), 776–785.

Wren, A.M., Seal, L.J., Cohen, M.A. et al.(2001) Ghrelin enhances appetite andincreases food intake in humans. Journalof Clinical Endocrinology and Metabolism86, 5992–5995.

Yeager, K.K., Agostini, R., Nattiv, A. &Drinkwater, B. (1993) The female athletetriad: disordered eating, amenorrhea,osteoporosis. Medicine and Science inSports and Exercise 25, 774–777.

Young, N., Formica, C., Szmukler, G. et al.(1994) Bone density at weight-bearingand non weight-bearing sites in balletdancers: the effects of exercise,hypogonadism and body weight. Journalof Clinical Endocrinology and Metabolism78, 449–54.

Zeni-Hoch, A., Dempsey, R.L., Carrera,G.F. et al. (2003) Is there an associationbetween athletic amenorrhea andendothelial cell dysfunction? Medicineand Science in Sports and Exercise 35(3),377–83.

Zipfel, S., Seibel, M.J., Lowe, B. et al. (2001)Osteoperosis in eating disorders: a follow-up study of patients withanorexia and bulimia nervosa Journal ofClinical Endocrinology and Metabolism86(11), 5227–5233.

Williams, N.I. (2003) Lessons fromexperimental disruptions of themenstrual cycle in humans andmonkeys. Medicine and Science in Sportsand Exercise 35(9), 1564–1572.

Williams, N.I., Young, J.C., McArthur, J.W.et al. (1995) Strenuous exercise withcaloric restriction: effect on luteinizinghormone secretion. Medicine and Sciencein Sports and Exercise 27(10), 1390–1398.

Williams, N.I., Clark, K.L., McConnell,H.J., Matuch, A. & O’Connor, K.A.(2000) Menstrual irregularities anddisordered eating in female athletes:survey vs. follow-up physiologicalstudies. Medicine and Science in Sports and Exercise 32(5), S64.

Williams, N.I., Caston-Balderrama, A.,Helmreich, D.L. et al. (2001a)Longitudinal changes in reproductivehormones and menstrual cyclicity incynomolgus monkeys during strenuousexercise training: abrupt transition toexercise induced amenorrhea.Endocrinology 142, 2381–2389.

Williams, N.I., Helmreich, D.L., Parfitt,D.B., Caston-Balderrama, A. &Cameron, J.L. (2001b) Evidence for acasual role of low energy availability in the induction of menstrual cycledisturbances during strenuous exercisetraining. Journal of Clinical Endocrinologyand Metabolism 86, 5184–5193.

Wilmore, J.H. (1991) Eating and weightdisorders in the female athlete.International Journal of Sports Nutrition 2,104–117.

Wilmore, J.H., Wambsgans, K.C., Brenner, M. et al. (1992) Is there energy

Page 295: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

279

Introduction

The mammalian testis is the site of germ cell develop-ment and androgen production (Rommerts 2004).Testosterone, a 19-carbon steroid secreted by thetestis, is the predominant androgen in most mam-mals. Testosterone plays a critical role in mam-malian reproduction; it is essential for maintainingsexual function, germ cell development and access-ory sex organs. In the adult animal, testosterone has additional effects on the muscle, bone, hema-topoeisis, coagulation, plasma lipids, protein andcarbohydrate metabolism, psychosexual and cog-nitive function. During sexual differentiation of the mammalian fetus, testosterone masculinizes theWolffian structures and causes the external genitaliato form a scrotum and penis. In addition, increasingtestosterone levels during pubertal developmentpromote somatic growth and virilization of boys.

Androgen production by the testes is regulatedprimarily by pituitary luteinizing hormone (LH),while germ cell development requires the co-ordinated action of follicle-stimulating hormone(FSH) and high intratesticular testosterone con-centrations generated by the Leydig cells under theinfluence of LH (Rommerts 2004). Paracrine interac-tions between Sertoli and germ cells are also import-ant in the regulation of spermatogenesis, althoughthe precise role of Sertoli cell in regulation of germcell development is not fully understood.

Testicular function is regulated by a series of feed-back and feedforward mechanisms that operate atthe level of the hypothalamus, the pituitary, and thetestes (Fig. 21.1). Thus, LH and FSH secretion by the

pituitary is stimulated by pulsatile gonadotropin-releasing hormone (GnRH) secretion, and regulatedby negative feedbacks from gonadal hormones,including gonadal steroids, and inhibin and activin.

Gonadotropin-releasing hormonesecretion by hypothalamic neurons

Developmental migration of GNRH neurons. The neurons that secrete GnRH originate in the region of the olfactory apparatus (Schwanzel-Fukuda &Pfaff 1989) and migrate, along the olfactory andvomeronasal nerves, into the forebrain and then intotheir final location in the hypothalamus (Fig. 21.2).This orderly migration of GnRH neurons requiresthe co-ordinated action of direction-finding mole-cules, adhesion proteins such as the KALIG-1 geneproduct and fibroblast growth receptor, and enzy-mes that help the neuronal cells burrow their waythrough intercellular matrix. Mutations of any ofthese proteins could arrest the migratory processand result in GnRH deficiency. In a subset of patients,failure of this developmental migration of GnRHneurons into their final location in the hypothala-mus results in a clinical disorder called idiopathichypogonadotropic hypogonadism (IHH) that ischaracterized by GnRH deficiency and impairedgonadotropin secretion by the pituitary (Legouis et al. 1991).

Hypothalamus as the integrating center for the malereproductive axis. The hypothalamus serves as theintegrating center for the reproductive system andco-ordinates the regulatory signals from the higher

Chapter 21

Regulation of Testicular Function: Changesin Reproductive Hormones During Exercise,Recovery, Nutritional Deprivation and Illness

SHALENDER BHASIN

Page 296: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

280 chapter 21

centers and the feedback from the gonads (Knobil1980; Crowley et al. 1991). The hypothalamus re-ceives neural input from the central nervous systemthat reflects the effects of emotion, stress, light, olfact-ory stimuli, temperature and other environmentalstimuli. The feedback signals from the gonadsinclude steroid hormones (testosterone and estra-diol) and protein hormones (inhibins and actvins).

Regulation of LH and FSH by pulsatile GnRH secretion.GnRH is a major regulator of gonadotropin secre-tion, and increases LH and FSH secretion from pituit-ary cells both in vitro and in vivo. Pulsatile secretionof GnRH is essential for maintaining normal LH andFSH secretion from the pituitary (Belchetz et al.1978; Knobil 1980; Shupnik 1990; Crowley et al. 1991;Weiss et al. 1992). Continuous GnRH infusion oradministration of a long-acting GnRH agonist leadsto decreased LH and FSH secretion, a phenomenonreferred to as down-regulation (Fig. 21.3) (Belchetzet al. 1978; Knobil 1980). The pattern of GnRH signal(amplitude and frequency) is important in deter-mining the quantity and quality of gonadotropinssecreted (Belchetz et al. 1978; Haisenleder et al. 1988,1991; Kim et al. 1988a, 1988b; Yuan et al. 1988;Shupnik 1990; Weiss et al. 1992). Marked increase in GnRH pulse frequency also desensitizes thegonadotrope, resulting in decreased LH and FSHsecretion (Belchetz et al. 1978; Mercer et al. 1988;Shupnik 1990). The electrophysiologic activity ofhypothalamic GnRH neurons is linked to episodicGnRH secretion.

The transcription of LH-β gene is induced by pulsatile GnRH administration in vitro (Wierman et al. 1989; Shupnik 1990; Weiss et al. 1992). Con-tinuous infusion of GnRH up-regulates only the α-gene transcription, but not LH or FSH β-subunitgene transcription (Haisenleder et al. 1988). PulsatileGnRH administration also modifies polyadenyla-tion of LH subunit mRNA (Weiss et al. 1992). The frequency of GnRH stimulus is important indifferential regulation of LH-β and FSH-β genes(Haisenleder et al. 1988). Faster frequencies increaseα and LH-β, and slower frequencies FSH-β, lead-ing to speculation that alterations in GnRH pulse frequency may be one mechanism by which twofunctionally distinct gonadotropins can be regu-lated by a single hypothalamic-releasing hormone

Light and otherenvironmental cues

HypothalamusGnRH

Emotion

Stress balance

Energy

Olfactory

Pituitarygonadotrope

Germ cellcompartment

Sperm

Leydigcells→T

Sertolicells

Inhibin

LH FSH

+

++

−+ +

T

T E2

Fig. 21.1 A schematic diagram of the hypothalamic–pituitary–testicular axis. The hypothalamus serves as the integrating center for the male reproductive axis.Emotion, olfaction, energy balance, light and stress exerttheir effects on human reproduction through circuits that impinge on hypothalamic gonadotropin-releasinghormone (GnRH) secreting neurons. Pulsatile GnRHsecretion by hypothalamic neurons stimulates luteinizinghormone (LH) and follicle-stimulating hormone (FSH)secretion by the pituitary. LH binds to G-protein-coupledreceptors on the Leydig cells in the testis and stimulatesthe production of testosterone; high intratesticulartestosterone concentrations along with FSH are essentialfor initiating and maintaining spermatogenesis.Testosterone has a negative feedback effect on pituitaryLH secretion and hypothalamic GnRH secretion directlyand indirectly through its conversion to estradiol.Circulating glycoproteins secreted by the Sertoli cells,known collectively as inhibins, also regulate FSHsecretion. As receptors for FSH and androgens are locatedon the Sertoli cells, it is generally believed that thehormonal influences on germ cells are mediated via theSertoli cells.

Page 297: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 281

(Haisenleder et al. 1988). Continuous infusion ofGnRH or administration of a GnRH agonist leads toa decrease in LH-β mRNA levels but α mRNA levelsremain elevated (Haisenleder et al. 1988; Kim et al.1988a, 1988b; Yuan et al. 1988).

A great deal of information about the physiologyof GnRH secretion has emerged from examinationof the LH and FSH pulse patterns in normal menand women, and from GnRH replacement studiesin patients with idiopathic hypogonadotropic hypo-gonadism (IHH) (Urban et al. 1988; Crowley et al.1991). Studies in such patients with hypothalamicGnRH deficiency have indicated that GnRH pulsesat a dose of 25 ng·kg–1, given intravenously to men,can replicate normal pulsatile LH secretion in all itscharacteristics (Crowley et al. 1991). The peak GnRH

levels achieved after intravenous administration ofthis dose (500–1000 pg·mL–1) are similar to thoseobtained in the primates by direct sampling of thehypophyseal-portal blood (Crowley et al. 1991). Inmen with IHH, an interpulse interval of 2 h appearsoptimum (Crowley et al. 1991). Increasing the fre-quency of GnRH pulses leads to progressive de-crease in LH responsiveness to GnRH (Rebar et al.1976). Decreasing the pulse frequency or increasingthe interpulse interval increases the amplitude ofthe subsequent LH pulse. There is a linear relation-ship between the log of the dose of GnRH pulse andthe amount of LH, FSH and free α subunit secreted(Spratt et al. 1986; Whitcomb et al. 1990). In the adultman, the magnitude of the LH response to GnRH isconsiderably greater than that of FSH.

11E

13E

14E

16E

vno

vno

gtob

vno

vno

ob

ob

gt

gt

poa

poa

Fig. 21.2 Developmental origin and migration of the gonadotropin-releasing hormone (GnRH) secreting neurons. Theneurons that secrete GnRH originate in the region of the olfactory apparatus. This figure shows the migratory route ofGnRH secreting neurons in sagittal sections of the mouse embryo as determined by GnRH immunohistochemistry. Onday 11, GnRH neurons are seen in proximity to the vomeronasal organ (vno) and the olfactory placode. By day 13, themigration of these neurons into the nasal septum and along the path of vomeronasal nerves and nervous terminalis isevident. By day 16, most of the GnRH neurons have entered the forebrain and some have entered into the preoptic area of the hypothalamus. gt, ganglion terminale; ob, olfactory bone; poa, preoptic area; vno, vomeronasal organ. (Reproducedfrom Schwanzel-Fukuda & Pfaff 1989.)

Page 298: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

282 chapter 21

1 Pulse/h

15 15 25 3510 10

Days

20 30 405 50

5 Pulse/h 1 Pulse/h40

35

30

25

20

15

10

5

0

LH (

ng

·mL–1

)

400

350

300

250

200

150

100

50

0

FSH

(n

g·m

L–1)

Pulsatile

15 25 3510 10

Days

20 305 50

Continuous Pulsatile

20

15

10

5

0

LH (

ng

·mL–1

)

200

150

100

50

0

FSH

(n

g·m

L–1)

Fig. 21.3 Knobil’s pioneering experiments demonstrated that the pulsatile nature of gonadotropin-releasing hormone(GnRH) signal is essential for maintaining normal luteinizing hormone (LH) and follicle-stimulating hormone (FSH)output. Knobil et al. (1980) ablated GnRH secreting neurons in the hypothalamus to create a monkey model that wasdeficient in GnRH. These monkeys also underwent gonadectomy in order to remove the feedback influences of thegonads. In this gonadectomized, GnRH deficient monkey model, pituitary LH and FSH secretion was normalized bypulsatile administration of GnRH at a frequency of one pulse per hour. Continuous infusion of GnRH decreased LH and FSH secretion, a phenomenon referred to as down-regulation. Similarly, administration of GnRH at an increasedfrequency of five pulses per hour resulted in decreased LH and FSH secretion. Restoration of pulse frequency to one pulseper hour restored normal LH and FSH output. (Reproduced with permission from Knobil et al. 1980 and Belchetz et al.1978.)

Page 299: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 283

Intensive sampling in normal adult men andwomen reveals a wide spectrum of LH pulse char-acteristics (Urban et al. 1988). The median values forLH pulse parameters in men, reported in one suchrecent study (Urban et al. 1988) were as follows:interpulse interval 55 min; LH peak duration 40 min;LH pulse amplitude 37% of basal (1.8 mLU·mL–1

incremental). Wide variations in LH pulse para-meters in apparently healthy normal men and wo-men dictate a need for caution in interpretation ofmild deviations in LH pulse frequency and ampli-tude parameters. The sampling frequency and theparadigm used to quantitate pulse characteristicscan have significant impact on the false-negativeand false-positive rates and on the observed pulseparameters (Urban et al. 1988).

GnRH action on the gonadotrope is mediated viaits binding to specific membrane receptors leadingto aggregation of receptors and calcium-dependentLH release (Conn et al. 1981, 1982).

Gonadotropin secretion by pituitary

Functional anatomy and development of the pituitarygland. The bulk of immunocytologic evidence sug-gests that a single cell type within the pituitarysecretes both LH and FSH (Moriarty 1973; Kovacs et al. 1985). Gonadotropes, the cells that secrete LHand FSH, constitute about 10–15% of anterior pitu-itary cells (Moriarty 1973; Kovacs et al. 1985) and aredispersed throughout the anterior pituitary close to the capillaries. Gonadotropes are easily demon-strable in the fetal and prepubertal pituitary gland(Childs et al. 1981); however, their number is lowbefore sexual maturation. Castration leads to anincrease in the size as well as the number ofgonadotropes. Adenohypophyseal cells are derivedfrom a common multipotential stem or progenitorcell. Genetic analyses of mutations associated withdevelopmental disorders of the pituitary have re-vealed the molecular mechanisms of pituitary devel-opment and cell lineage determination (Ingraham et al. 1988; Scully & Rosenfield 2002). Co-ordinated,temporal expression of a number of homeodomaintranscription factors directs the embroyologicaldevelopment of the pituitary and its differentiatedcell types. Three homebox genes Lbx3, Lbx4 andTitf1 are essential for early organogenesis (Scully

& Rosenfield 2002). Cell specialization and pro-liferation of differentiated cell types requires theexpression of transcription factors, Pit1 and Prop1.Pit-1 has a POU-specific and a POU-homeo DNA-binding domain (Scully & Rosenfield 2002). TheProp1 gene encodes a transcription factor with a single DNA-binding domain. While Pit-1 mutationsare associated with deficiencies of growth hormone(GH), thyroid-stimulating hormone (TSH) and pro-lactin, mutations in Prop1 are associated with defi-ciencies of LH and FSH in addition to deficiencies ofGH, prolactin and TSH. Expression of the HESX1gene precedes expression of Prop-1 and Pit-1; muta-tions in this gene are associated with septo-opticdysplasia and panhypopituitarism (Parks et al.1999).

Biochemical structure and molecular biology of LH andFSH. The family of pituitary glycoprotein hormonesincludes LH, FSH, TSH and chorionic gonadotropin(CG) (Sairam 1983; Ryan et al. 1987; Gharib et al.1990). Each of these hormones is heterodimeric, consisting of an α and a β subunit. The primarystructures of the α subunits of LH, FSH, TSH andCG are nearly identical within a species; the biologicspecificity is conferred by the dissimilar β subunit.Significant homology between the two subunitssuggests that these subunits arose from a commonancestral gene. Individual subunits are not biolo-gically active; formation of the heterodimer is essen-tial for biologic activity. The subunits are linkedinternally by disulfide bonds; the location of the cys-teines is important in determining the folding andthe three-dimensional structure of the glycoprotein(Sairam 1983; Ryan et al. 1987; Gharib et al. 1990).The α subunit of LH contains two asparagine-linkedcarbohydrate chains, while the β-subunit chain con-tains one or two (Table 21.1) (Baenziger 1990). TheCG β subunit also contains O-linked oligosaccha-rides not found on the LH dimer (Cole et al. 1984).Although free uncombined α subunit is secreted by the pituitary into the circulation, it is generallybelieved that the free β subunit is not secreted to anysignificant degree via this route.

Emergence of CG as a separate gonadotro-pin occurred relatively recently during evolution(Kornfeld & Kornfeld 1976; Fiddes et al. 1984).Unlike LH, which is found in the pituitaries of a

Page 300: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

284 chapter 21

large number of species, CG is found only in the placenta of certain mammalian species such ashorses, baboons and humans (Fiddes et al. 1984).The α and β subunits of LH and FSH are encoded by separate genes (Fiddes et al. 1984). The LH–CG-β gene cluster in the human incorporates seven CG-like genes, including one which codes for thehLH-β gene (Fiddes et al. 1984). The general organ-ization of the LH-β gene four exons and three intronsis similar to other glycoprotein hormone β genes(Table 21.1).

Regulatory role of LH. Testosterone secretion byLeydig cells is under the control of LH. LH binds to G-protein-coupled receptors on Leydig cells and activates the cyclic adenosine monophos-phate (cAMP) pathway. The luteinizing hormone–chorinonic gonadotropin (LH–hCG) receptor shareshomology with other G-protein-coupled receptorssuch as rhodopsin, adrenergic, TSH and FSH recep-tors (Fig. 21.4) (McFarland et al. 1989; Sprengel et al.1990). These G-protein-coupled receptors are trans-membrane proteins that share a structural motif of seven membrane-spanning domains. The aminoterminus of the receptor protein constitutes theextracellular domain. The carboxy terminus con-sists of the seven membrane-spanning segmentsand a small cytoplasmic tail, and has several serinesand threonines that may serve as phosphorylationsites (McFarland et al. 1989; Sprengel et al. 1990).

LH increases the activity of the side-chain cleav-

age enzyme (Simpson 1979; Mori & Marsh 1982), acytochrome P450-linked enzyme that converts cho-lesterol to pregnenolone, the rate-limiting step intestosterone biosynthesis. LH increases the deliveryof cholesterol to the side-chain cleavage enzyme,thus increasing its capacity to convert cholesterol topregnenolone (Simpson 1979; Mori & Marsh 1982).A steroidogenesis acute regulatory protein (STAR)makes cholesterol available to the cholesterol side-chain complex and regulates the rate of testosteronebiosynthesis (Clark & Stocco 1996). Peripheral ben-zodiazepine receptor, a mitochondrial cholesterol-binding protein involved in cholesterol transport, ispresent in high concentration in outer mitochon-drial membrane, and has also been proposed as an acute regulator of Leydig cell steroidogenesis.The long-term effects of LH include stimulation of gene expression and synthesis of a number of key enzymes in the steroid biosynthetic pathway,including the side-chain cleavage enzyme, 3-βhydroxysteroid dehydrogenase, 17-α hydroxylaseand 17,20-lyase (Simpson 1979; Mori & Marsh 1982).Although LH also activates phopholipase C path-way, it is unclear if this pathway is essential for LH-mediated stimulation of testosterone production. Inaddition, insulin-like growth factor-I, insulin-likegrowth factor binding proteins, inhibins, activins,transforming growth factor-β, epidermal growthfactor, interleukin-1, basic fibroblast growth factor,gonadotropin-releasing hormone, vasopressin andanother group of poorly characterized mitochondrial

Table 21.1 General structure of the pituitary glycoprotein hormones. There is considerable homology among theglycoprotein α, LH-β, FSH-β, TSH-β and hCG-β genes and proteins. (Reproduced from Grossman et al. 1997.)

Gene No. of mRNA No. of No. of glycosylation sites Subunit Locus length (Kb) exons (introns) length (Kb) amino acids (location)*

Common α 6p21.1-23 9.4 4 (3) 0.8 92 2 (N: 52,78)†

TSH-β 1p22 4.9 4 (3) 0.7 118‡ 1 (N: 23)LH-β 19q13.3 1.5 4 (3) 0.7 121 1(N: 30)CG-β 19q13.3 1.9§ 4 (3) 1.0 145 6 (N: 13,30; S: 121,127,132,138)FSH-β 11p13 3.9 4 (3) 1.8 111 2 (N: 7,24)

*Oligosaccharide chains are attached either to asparagine (N) (N-linked) or to serine (S) (O-linked). N or S residues arenumbered according to their position in the respective sequence.†Free subunit may also contain an additional site of O-glycosylation at threonine (T) 39.‡118-Amino acid coding region; six amino acids can be cleaved at the C-terminal end.§In contrast to all other glycoprotein hormone subunit genes which exist as a single copy, hCG is encoded by a cluster ofsix genes which vary in length.

Page 301: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 285

proteins have been implicated in control of steroido-genesis in Leydig cells.

The regulatory role of FSH in the male mammal. FSH binds to specific receptors on Sertoli cells andstimulates the production of a number of pro-teins including inhibin-related peptides, transferrin,androgen-binding protein, androgen receptor and7-glutamyl transpeptidase. However, the role ofFSH in the regulation of the spermatogenic processremains poorly understood. The prevalent dogma isthat LH acts on Leydig cells to stimulate the pro-duction of high intratesticular levels of testosterone(Boccabella 1963; Steinberger 1971; Sharpe 1987).Testosterone then acts on spermatogonia and prim-ary spermatocytes leading the germ cells throughthe meiotic division. FSH is felt to be essential forspermiogenesis, i.e. the maturation process bywhich spermatids develop into mature spermato-zoa. However, evidence from experimental animalsand patients with IHH treated with gonadotropinssuggest that FSH plays a more complex role in main-taining quantitatively normal spermatogenesis.

In the rat and non-human primate, testosteronealone can maintain spermatogenesis when admin-istered shortly after hypophysectomy or stalk re-section (Marshall et al. 1983; Sharpe et al. 1988;Stager et al. 2004). However, if testosterone is given after a lapse of several weeks to months, it ismuch less effective in reinitiating spermatogenesis.Spermatogenesis maintained by treatment of hypo-physectomized male rodent or non-human primateby testosterone is qualitatively, but not quantitat-ively, normal (Marshall et al. 1983; Sharpe et al. 1988;Stager et al. 2004). Combination of testosterone andFSH is more effective than testosterone alone inreinitiating spermatogenesis (Stager et al. 2004).Thus, although LH alone can maintain or reinitiatespermatogenesis, FSH is required for quantitativelynormal sperm counts.

In men with prepubertal onset of both LH andFSH deficiency, LH or hCG alone is unable to initi-ate spermatogenesis (Bardin et al. 1969; Matsumotoet al. 1983, 1984; Finkel et al. 1985). On the otherhand, if gonadotropin deficiency is acquired afterthe patient has completed pubertal development,

I II III IV

133

398

542

568

577

564

581578574571

554

V VI VII

ExtracellularNH3

IntracellularCOOH

V VI

616M

R

R

S

I

I

D

A

MA

TDC M

CA

T

T

TA

A

A

P

T

IK

K K

KKI

I

I

I

I

YF

F

FFS

S

F

AAV

N

N

P KL

L

M

545

A

C

Fig. 21.4 General structure of theluteinizing hormone–chorinonicgonadotropin (LH–hCG) receptor.The normal amino acids are shown,but the activating mutations areindicated by black circles with lightletters, whereas the inactivatingmutations are indicated by graycircles with white letters. Thecorresponding codon numbers alsoare shown. Most activating mutationsoccur in the sixth transmembranedomain and third intracellular loop,whereas inactivating mutations occur in the extracellular domain,transmembrane domains V–VII and the third intracellular loop.Activating mutations includeAsp578Gly, Ile542Leu, Asp564Gly,Asp578Tyr, Cys581Arg, Met571Ile,Thr577Ile, Ala568Val, Ala572Val andMet398Thr. Inactivating mutationsare Cys545X, Ala593Pro, Arg554X,Ser616Tyr and Arg133Cys. An Xindicates a stop codon, which is TAA,TAG or TGA. (Reproduced fromLayman 1999.)

Page 302: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

286 chapter 21

LH or hCG alone can reinitiate qualitatively normalspermatogenesis (Finkel et al. 1985). Thus, FSH isrequired for initiating the spermatogenic processbut, once this has occurred, testosterone in highdoses can maintain spermatogenesis. These observa-tions suggest that FSH may be involved in some sortof ‘programming’ in the peripubertal period, afterwhich LH alone can maintain germ cell develop-ment and maturation.

Androgen concentrations in the testis are muchhigher than those in serum. However, considerableconfusion exists with regard to the significance ofhigh intratesticular testosterone levels (Sharpe 1987;Sharpe et al. 1988; Stager et al. 2004). For example,stimulatory effects of exogenous testosterone onspermatogenesis in the rat are not associated withproportionate elevations of intratesticular testo-sterone levels. In the adult hypophysectorized orGnRH antagonist-treated monkey that is supple-mented with testosterone, a direct relationshipbetween testicular testosterone levels and sper-matogenesis has not been found (Stager et al. 2004). The method of post-mortem collection of testiculartissue affects estimation of intratesticular testo-sterone concentrations (Stager et al. 2004). Thus, the relationship between intratesticular testosteroneconcentrations, FSH and spermatogenesis remainspoorly understood. Androgen receptors are presenton Sertoli and peritubular cells, some Leydig cellsand endothelial cells of the small arterioles. How-ever, we do not know whether androgen receptorsare also present on germ cells. It is generally believedthat androgen effects on spermatogenesis are medi-ated indirectly through Sertoli cells, although it ispossible that testosterone might also directly affectgerm cell development. Testosterone affects proteinsecretion by both round spermatids and Sertolicells. The expression of androgen receptors is max-imal in Stages VI–VII of the seminiferous epithe-lium; testosterone regulates germ cell apoptosis in astage-specific manner.

The transduction of the FSH signals to the germcells requires mediation of Sertoli cells, as FSHreceptors are present on the Sertoli cells but are lacking on the germ cells. FSH receptor is also a G-protein-linked polypeptide consisting of a glycosy-lated extramembranous domain which is connected

to its C-terminus containing seven transmembranesegments (Sprengel et al. 1990).

Feedback regulation of luteinizinghormone and follicle-stimulatinghormone secretion

Feedback regulation by testosterone. Testosterone plays an important role in feedback regulation ofgonadotropins in the male. Serum LH levels risepromptly and serum FSH gradually after castrationin a number of experimental animals (Yamamoto et al. 1970; Badger et al. 1978). The mRNAs for α, LH-β (Gharib et al. 1986) and FSH-β (Gharib et al. 1987)increase after castration, although the changes inFSH-α mRNA are more modest. The postcastra-tion rise in serum LH and LH-β mRNA levels isbrought about both by an increase in the gona-dotrope number and hypertrophy of individualgonadotropes (Childs et al. 1987). Testosteronereplacement, started at the time of, or soon after,castration can attenuate the postcastration rise in αand LH-β mRNAs and serum LH levels, but has little effect on FSH-β mRNA levels (Gharib et al.1986, 1987).

The effects of testosterone on FSH secretion andsynthesis are complex. The net in vivo effect oftestosterone administration to normal men is inhibi-tion of serum FSH levels (Swerdloff et al. 1979;Winters et al. 1979). However, the direct effects of testosterone on FSH output at the pituitary levelare stimulatory (Steinberger & Chowdhury 1977;Bhasin et al. 1987; Gharib et al. 1987). In isolated pituitary cell cultures, testosterone increases FSHrelease into the media (Steinberger & Chowdhury1977). This is attended by a three to fourfoldincrease in FSH-β mRNA levels (Gharib et al. 1990).In intact male rats, in whom GnRH actions areblocked by administration of a GnRH antagonist,testosterone increases serum FSH levels in a dose-dependent manner (Bhasin et al. 1987). Bhasin et al.(1987) demonstrated that in castrated animalstreated with a GnRH antagonist, graded doses oftestosterone increase serum FSH levels. These dataindicate that the stimulatory effects of testosteroneon serum FSH levels are not mediated througheffects on a gonadal inhibitor of FSH, but rather

Page 303: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 287

directly at the pituitary level. Testosterone increasesFSH-β but not LH-β mRNA levels. However in theintact male animal, testosterone inhibits GnRH-stimulated FSH secretion, accounting for the netinhibition of serum FSH levels.

Testosterone inhibits LH secretion when given tonormal men and rats (Santen 1975; Matsumoto et al.1984; Veldhuis et al. 1984). These inhibitory effectsare largely exerted at the hypothalamic level, a con-clusion supported by observations that testosteronedecreases the frequency of LH pulses in eugonadalmen (Matsumoto & Bremner 1984; Scheckter et al.1989; Finkelstein et al. 1991a). Androgens have nodirect effects on LH-β mRNA levels in rat pituitarymonolayer cultures. Similarly, in GnRH antagonist-treated male rats, graded doses of testosterone leadonly to an increase in FSH-β but not LH-β mRNAlevels (Bhasin et al. 1987). In contrast to rats, in menwith IHH, the amplitude of LH pulses, initiated andmaintained by pulsatile GnRH therapy, is reducedby testosterone administration, indicating that inhumans, testosterone has additional actions at thepituitary level in attenuating pituitary LH responseto GnRH (Matsumoto et al. 1984; Scheckter et al.1989; Finkelstein et al. 1991a). These studies demon-strate that testosterone or one of its metabolitesinhibits gonadotropin secretion at both the pituitaryand hypothalamic levels in men.

The inhibitory effects of testosterone are medi-ated directly by testosterone itself and indirectlythrough its metabolites, estradiol and dihydro-testosterone (DHT). Administration of inhibitors of aromatase or 5-α reductase is associated withincreases in LH concentrations consistent with the role of estradiol and DHT in augmenting theinhibitory feedback effects of testosterone (Santen1975; Finkelstein et al. 1991b; Gormley & Rittmaster1992). However, administration of a non-aromatiz-able androgen, DHT, also inhibits LH secretion consistent with the proposal that aromatization of testosterone is not obligatory for mediating itsinhibitory effects on LH secretion (Santen 1975).Similarly, 5-α reduction of testosterone is not essen-tial for the LH-inhibitory effects of testosterone(Gormley & Rittmaster 1992). The hypothalamiceffects of testosterone are mediated via opiatergicpathways (Veldhuis et al. 1984).

Feedback inhibition by estrogens. Estrogens can exertboth stimulatory and inhibitory effects on gonado-tropin synthesis and secretion depending on thedose, duration, the presence or absence of GnRHand other physiologic factors. Data from experi-mental animals suggest that the stimulatory effectsof estrogens on gonadotropin synthesis and secre-tion in vivo are exerted directly at the pituitary level,while the inhibitory effects are mediated at thehypothalamic level (Neill et al. 1977; Clarke &Cummins 1982; Zmeili et al. 1986; Saade et al. 1989;Shupnik et al. 1989; Yamaji et al. 1992). Estrogenadministration leads to a decrease in LH pulse fre-quency suggesting a hypothalamic site of action(Shupnik et al. 1989). Estradiol treatment of hypo-thalamic slices decreases GnRH mRNA expression(Hall & Miller 1986). Finally, transcription of allthree gonadotropin subunits is negatively regulatedby estradiol in vivo, even though the direct in vitroeffects on pituitary are stimulatory (Neill et al. 1977;Clarke & Cummins 1982; Saade et al. 1989). Estra-diol inhibits LH pulse amplitude in normal men and in GnRH-deficient men maintained on GnRH(Finkelstein et al. 1991b). These studies provide evidence that in men, estradiol inhibits LH by anaction predominantly at the pituitary site.

Inhibins, activins and follistatins. The hypothesis that a peptide of gonadal origin selectively regulatesFSH secretion was postulated over 70 years ago(McCullagh 1932); however, it wasn’t until 1985 thatthe structure of inhibin-related peptides was finallycharacterized (Ling et al. 1985; Burger & Igarashi1988; Vale et al. 1988; Ying 1988). Inhibins aredimeric proteins consisting of a common α subunitand one of two β subunits, βA or βB (Fig. 21.5). Theheterodimers of α:βA form inhibin A and a:α:βB het-erodimers form inhibin B (Vale et al. 1988). InhibinsA and B are equipotent in their FSH-inhibitingpotency, although inhibin B appears to be the dominant circulating form of inhibin in men. Inaddition, βA subunits can form homodimers calledactivin A or heterodimers with the βB subunitscalled activin AB (Mason et al. 1985). Both activins Aand B stimulate FSH secretion in vitro.

Inhibin-related peptides are widely distributed inorgan systems and have significant homology with

Page 304: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

288 chapter 21

members of a family of proteins that includesMullerian inhibiting substance, transforming growthfactor-β, bone matrix proteins and the decapenta-plegic gene complex of Drosophila (Vale et al. 1988;Ying 1988), which play an important role as regula-tors of growth and differentiation in diverse tissues.Thus, activin interacts with erythropoietin to stimu-late erythropoiesis. Activin is also an important regulator of homeobox genes. In the testis, activinhas been shown to regulate spermatogonial multi-plication (Mather et al. 1990).

The role of inhibins in the adult male animalremains unclear. Immunoneutralization studies inrats reveal that infusion of anti-inhibin sera leads toan increase in serum FSH levels only in the femaleand prepubertal male animal, but not in the adultmale animal (Rivier et al. 1986). These studies ques-tioned the in vivo role of inhibin as an FSH regulatorin the adult male. Subsequently, Culler & Negro-Vilar (1990) demonstrated that when Leydig cells inadult male rats are destroyed by a Leydig cell-specifictoxin, ethane dimethane sulfonate (EDS), adminis-tration of anti-inhibin sera leads to an increase inserum FSH. These data suggest that under basalconditions in the adult male, testosterone plays amore important role in FSH regulation and thatinhibin effects are unmasked only when testosteronelevels are lowered. Indeed, using specific, two-sitedirected assays, an inverse relationship has beendemonstrated between circulating inhibin B levelsand FSH levels in healthy men and women, and inmen with disorders of germ cell development.

Although the original inhibin hypothesis postu-lated inhibin as a selective regulator of FSH, undersome conditions inhibins also regulate LH output(Vale et al. 1988). Conversely, both FSH and LH regulate inhibin production by Sertoli cells in the rat and in the human male (McLachlan et al. 1988;Keman et al. 1989; Krummen et al. 1989). FSH andLH both increase inhibin-α mRNA (Krummen et al.1989, 1990). FSH effects on inhibin subunits aremediated via cAMP (Najmabadi et al. 1993).

Follistatins are another class of FSH inhibitors(Ueno et al. 1987) that are made up of glycosylatedsingle chain polypeptides with homology to pancre-atic secretory inhibitory protein and human epider-mal growth factor. The mature follistatin proteincontains four repeating domains; three are highlysimilar among themselves and to hEGF and PSTI.The physiologic role of follistatins is not known;emerging data suggest that they may act primarilyto suppress FSH release. Follistatins are potentinhibitors of estrogen production in granulosa cellsand can bind activin. Follistatins also act as bindingproteins for other growth regulating proteins suchas myostatin.

Activins regulate intragonadal function in boththe male and the female (Vale et al. 1988). In thetestis, activins suppress LH-stimulated testosteroneproduction while inhibins suppress it. In granulosacells, activins increase aromatase activity but inhibitprogesterone synthesis (Vale et al. 1988; Ying 1988).Activin B may also act as an autocrine/paracrinemediator in the pituitary and modulate FSH-α geneexpression.

Ontogenesis of luteinizing hormone and follicle-stimulating hormonesecretion during different phases ofhuman life

Fetal life. GnRH is demonstrable in the fetal hypo-thalamus at 6 weeks of gestation (Lee, P.A. 1988).Fetal pituitary contains measurable amounts of LH and FSH by 10 weeks, and by 11–12 weeks LHresponse to GnRH can be shown. Serum LH andFSH levels rise to a peak at about 20 weeks(Clements et al. 1980; Lee, P.A. 1988). In the secondhalf of pregnancy, serum LH and FSH levels in the

α subunit

20 KDa

Inhibin A

βA subunit

15 KDa

Inhibin B

Activin A Activin BActivin AB

βB subunit

15 KDa

Fig. 21.5 General structure of the inhibin/activin familyof gonadal hormones.

Page 305: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 289

fetus decline gradually. The mechanism of thisdecline is not known but several factors operative inthe second half of pregnancy may be responsible.The rise in sex steroid secretion by the fetal gonad,the rising maternal estrogen levels and the develop-ment of negative feedback mechanisms may all contribute to tonic inhibition of the hypothalamusand the pituitary gland by the ambient sex steroidconcentrations (Winter et al. 1975; Sizonenko 1979).

Placental hCG plays a significant role in stimulat-ing androgen production by the fetal testis in earlypregnancy (Lee, P.A. 1988). High androgen levels arerequired for differentiation of Wolffian structures inthe male. In addition, FSH stimulates differentiationand development of seminiferous tubules. Thesedata are consistent with observations that patientswith IHH have normal differentiation of Wolffianstructures and external genitalia because the placen-tal hCG drives the fetal testis to produce sufficientandrogen, even in the absence of pituitary LH andFSH. However, because of FSH deficiency, thesepatients have arrested or retarded development of seminiferous tubules. Testicular descent is partlydependent, during the later part of pregnancy, onandrogen levels (Husmann 1991) which, during thisperiod of fetal life, are maintained by pituitary LH.

Postnatal life and childhood years. After birth, serumLH and FSH levels rise again, albeit transiently(Winter et al. 1975; Sizonenko 1979). In the first 6 months of postnatal life, LH and FSH levels aremeasurable in blood (Winter et al. 1975). In fact, thepulsatile pattern of LH and FSH secretion is easilydiscernible during this brief period of reactivationof the hypothalamic–pituitary axis (Winter et al.1976; Jakachi et al. 1982). Serum LH and FSH levelspeak around 2–3 months of age and then decline toundetectable levels by 9–12 months; serum testos-terone levels undergo similar changes. This briefperiod of postnatal life thus provides a window inwhich the normality of the hypothalamic–pituitary–gonadal axis can be assessed before gonadotropinand sex steroid levels fall back to the low range.

During childhood years, the hypothalamic–pituitary–gonadal unit is kept in abeyance until theonset of puberty (Lee, P.A. et al. 1976; Apter et al.1978). However, the pituitary and the testis retain

the ability to respond to GnRH and to hCG, respect-ively. The response of prepubertal pituitary toGnRH stimulus is relatively damped. In addition,the GnRH-induced rise in serum FSH in prepubertalhumans is greater than that in LH. This is in contrastto an adult individual in which a single dose ofGnRH causes a greater rise in LH. During pubertalmaturation, serum LH and FSH levels rise (Sizonenko1979). The activation of FSH secretion precedes thatof LH. Sleep-entrained pulsatile secretion of LH is highly characteristic of early stages of puberty( Jakachi et al. 1982).

The use of highly sensitive two site-directedimmunoradiometric, immunofluorometric and che-miluminescent assays has revealed that between 7 years of age and adulthood mean LH concentra-tions increase over a 100-fold, mean FSH concentra-tions sevenfold and estradiol levels 12-fold (Apter et al. 1989). The increase in FSH is gradual, but therise in LH is steep. The changes in FSH precede therise in LH concentrations. The mechanisms thatkeep GnRH secretion in check during childhoodand trigger GnRH secretion at the onset of pubertyremain unknown.

Changes in reproductive function during aging. There isagreement that serum testosterone levels declineprogressively in men with advancing age (Fig. 21.6)(Pirke & Doerr 1973; Dai et al. 1981; Gray et al. 1991;Simon et al. 1992; Zmuda et al. 1997; Ferrini &Barrett-Connor 1998; Harman et al. 2001; Feldman et al. 2002; Matsumoto 2002); almost 25% of menover the age of 70 have serum testosterone levels inthe hypogonadal range (Harman et al. 2001). As sexhormone binding globulin (SHBG) levels are higherin older men than in younger men, the decrease infree and bioavailable testosterone with aging isgreater than the decline in total testosterone levels(Pirke & Doerr 1973; Tenover et al. 1987; Ferrini & Barrett-Connor 1998; Harman et al. 2001). Thediurnal rhythm of testosterone secretion, observedin younger men, is attenuated in older men. Whilemean serum levels of total, free and bioavailabletestosterone fall in the later decades of life, manyelderly men retain serum testosterone in the normalmale range. There is either no change or a modestincrease in circulating estradiol and estrone levels

Page 306: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

290 chapter 21

with age due to the increased peripheral aromatiza-tion of androgen to estrogen (Pirke & Doerr 1973;Ferrini & Barrett-Connor 1998).

The age-related changes in reproductive hor-mones are compounded by the effects of conco-mitant illness, changes in body composition andmedications. Although the data on the relationshipof age to serum androgen levels are mostly cross-sectional in nature, a few longitudinal studies(Zmuda et al. 1997; Harman et al. 2001; Feldman et al.2002) have confirmed the aging-associated declinein serum testosterone levels. Some studies havebeen criticized for selecting elderly men who werehealthier than the general population. However,even after adjusting for illness, time of sampling,assay variability and medications, testosterone levels are lower in older men than in younger men.

Aging-associated decline in testosterone levelsoccurs due to defects at all levels of the hypothalamic–pituitary–gonadal axis. Androgen secretion by thetestis of elderly men is decreased due to primaryabnormalities at the gonadal level. This is supportedby their higher basal LH and FSH levels (Kaufman& Vermeulen 1997; Morley et al. 1997; Feldman et al.2002), decreased testosterone response to hCG and diminished Leydig cell mass in aging men(Longcope 1973). In addition, secondary defectsmay exist at the hypothalamic–pituitary level, asindicated by the somewhat blunted LH and FSHresponses of older men to GnRH (Harman et al.1982). The circulating levels of free α subunit arealso higher in older men as compared to youngermen (Harman et al. 1982). Older men secrete LHmore irregularly than younger men (Pincus et al.1997). The older men also have less synchronicity

between LH and testosterone secretion than youngermen (Pincus et al. 1997). Therefore, aging is asso-ciated with abnormalities of the normal feedbackcontrol mechanisms that control the flow of informa-tion between components of the hypothalamic–pituitary–testicular network, and a disruption of the orderly pattern of pulsatile hormonal secretion(Pincus et al. 1997).

Testosterone secretion, transport andmetabolism

Testosterone secretion. In males of most mammalianspecies, 95% of circulating testosterone is derivedfrom testicular secretion. In man, 3–10 mg of testo-sterone is secreted daily by the testis (Horton 1978).Direct secretion of testosterone by adrenal, and peri-pheral conversion of androstenedione, collectivelyaccount for another 500 µg of testosterone daily.Only a small amount of dihydrotestosterone (approx-imately 70 µg daily) is secreted by the human testis;most of circulating DHT is derived from peripheralconversion of testosterone (Longcope & Fineberg1985).

Testosterone is produced in the testis by a het-erogeneous group of cells that includes the adultLeydig cells, Leydig cell precursors and immatureLeydig cells (Prince 2001). Studies in hypogona-dotropic (hpg) mice suggest that fetal developmentof both Sertoli and Leydig cells is independent ofgonadotrophins; however, normal differentiationand proliferation of the adult Leydig cell populationrequires the presence of gonadotrophins. 46,XYmale humans with inactivating mutations of LHreceptor have varying degree of genital ambiguity

(177)(144) (151)

(158)

(109)(43)

30 40 50 60Age (years)(a)

70 80 90

20

18

16

14

12

10

Test

ost

ero

ne

(nm

ol·L

–1)

(177)

(144)

(151)

(158)

(109)

(43)

30 40 50 60Age (years)(b)

70 80 90

0.5

0.4

0.3

0.2

0.1

Free

T in

dex

(nm

ol·n

mo

l–1)

Fig. 21.6 Age-related changes in serum total testosterone levels (a) and free testosterone (T) index (b) in men participatingin the Baltimore longitudinal study of aging. (Reproduced from Harman et al. 2001.)

Page 307: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 291

and Leydig cell agenesis, pointing to the importantrole of LH in the regulation of Leydig cell develop-ment (Dufau 1988; Huhtaniemi & Toppari 1995). Sertoli cell number after birth is regulated bygonadotrophins.

Androgen transport in the body. Ninety eight percentof circulating testosterone is bound to plasma pro-teins: the SHBG and albumin (Vermeulen 1988;Rosner 1991). The SHBG binds testosterone withmuch greater affinity than albumin. Only 0.5–3.0%of testosterone is unbound. Although the preval-ent dogma assumes that only the unbound fraction is biologically active, albumin-bound hormone dissociates readily in the capillaries and may bebioavailable (Pardridge 1987). Pardridge (1987) hasdemonstrated that albumin- and SHBG-boundandrogens represent the major circulating pool ofbioavailable hormone for testis or prostate. Further-more, these investigators have argued that SHBG-sex steroid complex may be nearly completelyavailable for influx through the blood–testis barrieror prostate plasma membrane; this view is not universally shared (Pardridge 1987).

SHBG is a glycoprotein, synthesized in the liver,that possesses high affinity binding for testoster-one and estradiol (Vermeulen 1988; Rosner 1991).Hepatic production of SHBG is regulated by insulin,thyroid hormones, dietary factors and the balancebetween androgens and estrogens. SHBG is involvedin the transport of sex steroids in plasma, and itsconcentration is a major factor regulating their distribution between the protein-bound and freestates. Plasma SHBG concentrations are decreasedby androgen administration, obesity, hyperinsulin-ism and nephrotic syndrome (Rosner 1991). Con-versely, estrogen administration, hyperthyroidism,many types of chronic inflammatory illnesses andaging are associated with high SHBG concentra-tions. A locus that is associated with SHBG con-centrations in African-Americans and Caucasianshas been mapped to 1q44; several additional loci in African-Americans also exhibit linkage withSHBG concentrations, suggesting that many genesregulate SHBG levels (Larrea et al. 1995). The binding of testosterone to SHBG or albumin is notessential for androgen action; rats that are deficient

in SHBG and albumin are fertile and have normalmating behavior.

Testosterone metabolism. Testosterone is metabolizedpredominantly in the liver (50–70%) although somedegradation also occurs in peripheral tissues, par-ticularly the prostate and the skin. Liver takes up testosterone from the blood and through a seriesof chemical reactions, which involve 5-α- and 5-β-reductases, 3-α- and 3-β-hydroxysteroid dehydro-genases and 17-β-hydroxysteroid dehydrogenase,converts it into androsterone, etiocholanolone, bothinactive metabolites, and dihydrotestosterone and3-α-androstanediol. These compounds undergo glu-coronidation or sulfation before their excretion by the kidneys. Free and conjugated androsteroneand etiocholanaolone are the predominant urinarymetabolites of testosterone.

Testosterone as a prohormone: the role of DHT and estra-diol in mediating androgen action. Testosterone is con-verted in many peripheral tissues into its activemetabolites, estradiol 17-β and 5-α-DHT (Wilson et al. 1993; Grumbach & Auchus 1999). Aromatiza-tion of the A ring converts it into 17-β-estradiol. In addition, reduction of the δ-4 double bond canconvert testosterone into 5-α-DHT. Testosteroneactions in many tissues are mediated through thesemetabolites. For instance, testosterone effects on trabecular bone resorption, sexual differentiation of the brain, plasma lipid concentrations, atheros-clerosis progression and some types of behaviorsare mediated through its conversion to estrogen(Grumbach & Auchus 1999). The study of mice that have mutantions in the estrogen receptor-α,estrogen receptor-β, or the aromatase gene has pro-vided considerable insight into the role of estrogenin male mammals (Jones et al. 2000). These models ofestrogen deficiency exhibit significant disruption of spermatogenesis and fertility, elevated testosterone and LH levels, decreased bone mass and increasedadiposity, indicating the important role of estrogensin regulation of bone mass, gonadotropin regula-tion, body composition and spermatogenesis (Smithet al. 1994; Carani et al. 1997). A small number ofhumans with inactivating mutations of the CYP19aromatase gene have been reported (Carani et al.

Page 308: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

292 chapter 21

1997). Females with CYP19 gene mutations are mas-culinized, fail to undergo pubertal development,have elevated levels of androgens and LH and FSH,polycystic ovaries and tall stature. Males with CYP19aromatase mutations are characterized by osteo-porosis, increased bone turnover, delayed epiphy-seal fusion, tall stature and increased testosteronebut decreased estradiol levels (Carani et al. 1997).

Two isoenzymes of steroid 5-α-reductase havebeen characterized (Wilson et al. 1993; Russell &Wilson 1994). Type 1 steroid 5-α-reductase isexpressed in many non-genital tissues, has beenmapped to chromosome region 5p15 and has a pHoptimum of 8. Type 2 steroid 5-α-reductase isexpressed in the prostate and other genital tissues,has been mapped to 2p23 and has a pH optimum of 5.0 (Wilson et al. 1993). The biological role of 5-α-reductase type 1 has not been ascertained fully.The gene-targeting experiments suggest that type 1 enzyme plays a role in progesterone metabolism at the end of pregnancy (Wilson et al. 1993). Themice lacking 5-α-reductase enzyme type 1 have fail-ure of cervical ripening and fail to deliver (Wilson et al. 1993).

Testosterone effects on the prostate and seba-ceous glands of the skin require its 5-α reduction toDHT. DHT has been implicated in the pathophysio-logy of benign prostatic hyperplasia and androgenicalopecia (Wilson et al. 1993). Type 2 isoenzyme is the predominant form in the prostate, and has been implicated in the pathophysiology of benignprostatic hypertrophy, hirsutism and possibly male-pattern baldness. During embryonic life, testo-sterone controls the differentiation of the Wolffianducts into epididymis, vasa deferentia and seminalvesicles. The development of structures from theurogenital sinus and the genital tubercle such as the scrotum, penis and penile urethra require theaction of DHT. Although testosterone and DHTboth exercise anabolic effects on the muscle, steroid5-α-reductase activity is very low or absent in theskeletal muscle, and we do not know whether 5-αreduction of testosterone to DHT is obligatory formediating androgen effects on the muscle. Sim-ilarly, the literature is unclear on whether androgeneffects on sexual function in adult men are mediatedthrough testosterone or DHT.

A large body of information about the role ofDHT has emerged from the study of patients withautosomal recessive, steroid 5-α-reductase defi-ciency. 46,XY males with this syndrome contain normal male internal structures including testes,but exhibit ambiguous or female external genitaliaat birth (Cai et al. 1996; Mendonca et al. 1996). Atpuberty, these individuals undergo partial viriliza-tion and normal muscular development (Cai et al.1993). Many, but not all, 46,XY individuals with thisdisorder develop male gender identity, even if theyhave been brought up as females. Their develop-ment suggests that testosterone itself is able to stimulate psychosexual behavior, libido, develop-ment of the embryonic Wolffian duct, muscle devel-opment, voice deepening, spermatogenesis andaxillary and pubic hair growth. In contrast, DHT isrequired for prostate development and growth, the development of the external genitalia and male patterns of facial and body hair growth or male-pat-tern baldness. All 5-α-reductase-deficient kindreds,studied to date, have been shown to have mutationsin steroid 5-α-reductase type 2, the predominantform in the prostate (Cai et al. 1996; Mendonca et al.1996).

Mechanism of androgen action. Most actions of testo-sterone and DHT are mediated through binding to an intracellular androgen receptor that acts as aligand-dependent transcription factor (Zhou et al.1994; Lee, D.K. & Chang 2003). Testosterone bindsto the androgen receptor with half the affinity ofDHT, although the maximal binding capacity issimilar for both androgens. The DHT–androgenreceptor complex has greater thermostability and a slower dissociation rate than the testosterone-receptor complex. This may confer greater potencyto DHT in mediating androgen effects in someandrogen sensitive tissues, such as the prostate.

The androgen receptor has homology to othernuclear receptors, including the receptors for gluco-corticoids, progesterone and mineralocorticoids(Zhou et al. 1994; Lee, D.K. & Chang 2003). The pre-dominant 919-amino acid, 110–114 kDa, androgen receptor protein has three conserved functionaldomains: the steroid binding domain, the DNAbinding domain and the transcriptional activational

Page 309: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 293

domain; of these, the central, cysteine-rich DNAbinding domain is the most conserved. The singlecopy androgen receptor gene spans a 90 kb regionon the chromosomal region Xq11–12. In the absenceof its ligand, the androgen-receptor protein is dis-tributed both in the nucleus and the cytoplasm.However, androgen binding to the receptor causesit to translocate to the nucleus; amino acid sequencesbetween 617–633 of the androgen receptor areimportant for its nuclear migration and transac-tivation function. There is inconclusive evidencethat some androgen effects may be mediatedthrough nongenomic mechanisms through mem-brane receptors.

The binding of androgens by the androgen recep-tor results in conformational changes in this protein;there is evidence that the binding of anti-androgensto the androgen receptor might induce a differentset of conformational changes (Zhou et al. 1994; Lee,D.K. & Chang 2003). The androgen receptor can usetwo transactivation domains, AF1 and AF2, respect-ively. The transactivation domain AF1 (includingthe so-called 1 and 5 regions) is located in theaminoterminal part of the receptor, whereas AF2 islocated in the carboxyterminal, ligand-dependentdomain. In the intact receptor, both AF1 and AF2 areligand-dependent and influenced by nuclear recep-tor coactivators. In contrast, in a truncated androgenreceptor, that is missing the ligand-binding domain,AF1 becomes constitutively active. Hormone bind-ing to the androgen receptor results in assembly of tissue-specific co-activators and co-repressors thatdetermine the specificity of hormone action.

The mutations of the androgen-receptor genehave been associated with a wide spectrum of phenotypic abnormalities (Brinkmann 2001). Pati-ents with complete androgen insensitivity presentwith male pseudohermaphroditism, characterizedby female external genitalia, a blind vaginal pouchand well-developed breasts. Other patients withandrogen-receptor mutations may have a male phe-notype and milder abnormalities such as hypospa-dias, gynecomastia and infertility (McPhaul et al.1993).

The lengths of the CAG and GCG repeats in exon1 of the androgen-receptor gene have been linked totranscriptional activity of androgen-receptor pro-

tein. An abnormal length of the polyglutamine tractin exon 1 of the androgen receptor has been asso-ciated with spinal and bulbar muscular atrophy,also known as Kennedy’s disease. Although somestudies have reported an association of polymor-phisms in the lengths of polyglutamine and poly-glycine tracts with male infertility and risk of prostatecancer, others have not confirmed these findings(Casella et al. 2001).

The biochemical pathways that linkenergy balance and reproductive axis

Humans have known since antiquity that energybalance and nutritional status are intimately linkedto the reproductive axis in both men and women.The onset of puberty, the length of the reproductiveperiod, the number of offspring and the age ofmenopause have all been linked to body weight and composition, particularly the amount of bodyfat (Frisch & McArthur 1974; Van Der Spruy 1985;Frisch 1989; Foster & Nagatani 1999). Normal repro-ductive function requires an optimal nutritionalintake; both caloric deprivation and consequentweight loss, and excessive food intake and obesityare associated with impairment of reproductivefunction. The temporal aspects of sexual maturationare more closely associated with body growth thanwith chronological age (Penny et al. 1978; Frisch1989). In the animal kingdom, during periods offood scarcity, small animals with a short-life spanmay not even achieve puberty before death (Foster& Nagatani 1999). In animals with longer life spans,sexual maturation may be delayed during fooddeprivation. Undernutrition, caused by famine, eating disorders and exercise, results in weight lossand changes in body composition and endocrinemilieu that can impair reproductive function(Penny et al. 1978; Bates et al. 1982; Rock et al. 1996).As a general rule, weight loss and body compositionchanges resulting from undernutrition are asso-ciated with reduced gonadotropin secretion; thedecrease in FSH and LH levels correlates with thedegree of weight loss (Fig. 21.7) (Penny et al. 1978).However, both hypogonadotropic and hyper-gonadotropic hypogonadism have been describedin cachexia associated with chronic illnesses, such as

Page 310: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

294 chapter 21

ExerciseExercise

energy drain

Bodyweight andcomposition

ImpairedGnRH

secretion

StarvationEating

disorders

IllnessStress

Impaired reproductive function:decreased sex steroid secretion;

impaired fertility; delayed puberty;menstrual irregularities

(a)

NPY

Leptin

LeptinPituitary

NOSNO

Leptinreceptor

GnRH

GALP+

+

+

+

NPY++

+

+ −

POMC

Foodintake

Energyexpenditure

StressIllness

LH & FSH

Adipocytesenergystores

(b)

Central controlof reproduction

Central controlof energy balance

CRH

Fig. 21.7 Relationship betweenenergy balance, exercise, illness and gonadotropin secretion. (a)Endocrine signals that emanate fromadipocytes and are integrated in thehypothalamus and other centralsensors regulate hypothalamicgonadotropin-releasing hormone(GnRH) secretion. In addition,products and mediators of systemicinflammatory and stress responsesduring illness and exposure tophysical, chemical or psychologicalstress affect the male reproductiveaxis at multiple levels, resulting inimpairment of sex steroid secretion,pubertal development and fertility.(b) Hypothetical depiction of thebiochemical pathways that linkenergy balance and reproductiveaxis. The prevalent hypothesis is thatmetabolic signals that link energystores to hypothalamic GnRHsecretion originate in the adipocytesand are communicated to thehypothalamic GnRH secretingneurons through leptin, GALP andother poorly understood chemicalsignals. Leptin, the product of the obesity gene (ob) inhibitsneuropeptide Y (NPY) which has a tonic inhibitory effect on GnRHsecretion. Leptin also has directstimulatory effect on luteinizinghormone (LH) secretion bystimulating nitric oxide (NO)production in the gonadotropes.Therefore, the net effect of leptinaction is stimulation of LH secretion.Leptin decreases food intake whileother mediators of stress response,including products of the POMCgene and corticotropin-releasinghormone, inhibit food intake. FSH,follicle-stimulating hormone; NOS,nitric oxide synthase. (Reproducedfrom Bross et al. 2000.)

Page 311: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 295

human immunodeficiency virus (HIV) infection(Arver et al. 1999). Collectively, these observationsprovide compelling evidence that energy balance isan important determinant of reproductive functionin all mammals.

We do not know the precise nature of the bio-chemical pathways that connect energy metabolismand the reproductive axis, two biologic systemsessential for the survival of all species. The preval-ent hypothesis is that the metabolic signals, that re-gulate hypothalamic GnRH secretion, are mediatedthrough leptin and neuropeptide Y (Aubert et al.1998; Clarke & Henry 1999; Cunningham et al. 1999;Foster & Nagatani 1999). Leptin, the product of theobesity (ob) gene, is a circulating hormone secretedby the fat cells that acts centrally to regulate theactivity of central nervous system effector systemsthat maintain energy balance (Schwartz et al. 1999).Leptin stimulates LH secretion by activation of thenitric oxide synthase in the gonadotropes (McCannet al. 1998), and inhibits neuropeptide Y secretion.Neuropeptide Y has a tonic inhibitory effect on bothleptin and GnRH secretion. Leptin also stimulatesnitric oxide (NO) production in the mediobasalhypothalamus; NO stimulates GnRH secretion bythe hypothalamic GnRH-secreting neurons (McCannet al. 1998). More recent evidence suggests that additional pathways including those that involve agalanin-like peptide (GALP) -link energy homeo-stasis, food intake and GnRH secretion in the hypo-thalamus (see Fig. 21.7) (Seth et al. 2004). The neteffect of leptin action is stimulation of hypothalamicGnRH secretion (Schwartz et al. 1999).

Caloric deprivation in mammals is associatedwith reduced leptin levels and a concomitant reduc-tion in LH levels (Schwartz et al. 1999). Leptinadministration to calorically-deprived mice reversesthe inhibition of gonadotropin secretion that attendsfood-restriction (Schwartz et al. 1999). Similarly,genetically ob/ob mice with leptin deficiency havehypogonadotropic hypogonadism and are infertile;treatment of these mice with leptin restores gona-dotropin secretion and fertility (Mohamed-Ali et al.1998; Schwartz et al. 1999). Thus, energy deficit andweight loss are associated with impaired GnRHsecretion; in part, because of changes in neuro-peptide Y and GALP activity and the consequent

decrease in leptin secretion. While leptin is an important metabolic signal that links energy bal-ance and the reproductive axis, it remains unclearwhether it is the primary trigger for the activation ofthe GnRH-pulse generator at the onset of puberty.Emerging evidence suggests that leptin is essentialbut not sufficient for the initiation of puberty.

Historical and experimental illustrationsof the link between nutritional status,reproduction and fertility

The Dutch Hunger Winter. Between October 1944 andMay 1945, during the course of the Second WorldWar, German army restricted food supplies in certain Dutch cities (Fig. 21.8). This resulted in sub-stantial reduction in average daily energy intake toless than 1000 kcal (Stein et al. 1973) in cities affectedby the German siege. In some adjacent cities, foodsupplies were not curtailed by the Germans (con-trol cities). Studies by Stein et al. (1973) revealed that 50% of women, affected by famine, developedamenorrhea, the conception rate dropped and therewas increased perinatal mortality, congenital mal-formations and schizophrenia. Thus, optimal caloricintake is essential for normal fertility and prenatalgrowth.

The association of weight change and fertility in the!Kung San of Botswana. The !Kung San of Botswanawere a tribe of hunter–gatherers (Fig. 21.9) (Van DerWalt et al. 1978). The body weight of men andwomen in the tribe varied throughout the yeardepending upon food availability. In summermonths, when food supply was abundant, bodyweight increased, while the nadir of body weightwas achieved in winter months. The number ofbirths in the tribe peaked about 9 months after thepeak of body weight (Van Der Walt et al. 1978). Thisis another example of how the availability of foodregulates fertility patterns in nature.

The Minnesota caloric deprivation experiment. In thelate 1940s, Ancel Keys and coworkers studiedhuman starvation in an experiment in which 32young men volunteered to live on the campus of

Page 312: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

296 chapter 21

Eindhoven

UtrechtMass R

Mass R

Mass R

Leiden

Famine city above 500 000 populationFamine city 100 000–500 000 populationFamine city 40 000–100 000 populationControl city 100 000–500 000 populationControl city 40 000–100 000 population

The Hague

Hearlem

Amsterdam

Rotterdam

Groningen

NorthSea

0 km

4000

Ave

rag

e d

aily

cal

ori

c in

take

Bir

ths

3500

3000

2500

2000

1500

1000

J0

100

TilburgBreda

Swolle

Maastricht

Mangolo

Enachade

Hearlen

(a)

(b)

Leeuwarden

FMAM J J A S OND J FMAM J J A S OND J FMAM J J A S OND

8000Totalbirths

Conceivedduring famineJuly '46-Feb '48

Born duringfamine

Oct '44-May '48

Calorieration atconception

7000

6000

5000

4000

3000

2000

Fig. 21.8 The Dutch Hunger Winterduring the German siege. During theGerman siege of the Netherlandsduring the Second World War,residents of many Dutch citiesexperienced severe curtailment of caloric intake (famine cities).Adjacent cities that did not facecurtailment of food supplies wereused as control cities (a). (b) Therelationship between caloric intake and the number of births.(Reproduced from Stein et al. 1973.)

Page 313: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 297

the University of Minnesota and consume a dietproviding approximately 1600 kcal·day–1 (6.688MJ·day−1), about two thirds of their normal energyrequirement (Keys et al. 1950). The volunteers lost an average of 23% of their initial body weight;more than 70% of body fat and 24% of lean tissue.Decreased caloric intake and subsequent weightloss first caused a loss of libido and a reduction inprostate fluid, sperm motility and longevity. Sperm

production was reduced when men weighed ~ 25%less than the normal weight for their height. Weightgain restored reproductive function.

Reproductive dysfunction in acute andchronic illnesses

There is a high prevalence of androgen deficiencydefined solely in terms of low testosterone levels in men with chronic illness. Thus, even after theadvent of potent anti-retroviral therapy, androgendeficiency continues to be a common complicationof HIV infection in men. In an earlier survey of 150 HIV-infected men who attended our HIV clinicin 1997, approximately one third had serum total and free testosterone levels in the hypogonadalrange (Arver et al. 1999). Other investigators havereported a similar prevalence of hypogonadism inHIV-infected men (Dobs et al. 1996; Grinspoon et al.1996). A recent survey of HIV-infected men foundthe prevalence of low testosterone levels to be 20%(Reitschel et al. 2000). Thus, androgen deficiencycontinues to be a common occurrence in HIV-infected men.

In our survey, 20% of HIV-infected men with lowtestosterone levels had elevated LH and FSH levels,and thus had hypergonadotropic hypogonadism(Arver et al. 1999). These patients presumably hadprimary testicular dysfunction. The remaining 80%had either normal or low LH and FSH levels. Themen with hypogonadotropic hypogonadism eitherhad a central defect at the hypothalamic or pituitarysite or a dual defect involving both the testis and thehypothalamic–pituitary centers. The pathophysio-logy of hypogonadism in HIV infection is com-plex and involves defects at multiple levels of thehypothalamic–pituitary–testicular axis.

In a recent study, a majority of men with chronicobstructive lung disease had low total and freetestosterone levels (Casaburi et al. 1996). Similarly,there is a high frequency of hypogonadism in pati-ents with cancer, end-stage renal disease on hemo-dialysis and liver disease (Handelsman & Dong1993; Singh et al. 2001). Previous reports suggest that two thirds of men with end-stage renal diseasehave low total and free testosterone concentrations(Handelsman & Dong 1993). In a recent study, of the

Nu

mb

er o

f b

irth

s ( —

)50

45

40

35

30

25

20

15

10

Bo

dy w

eigh

t in Lb

s (…)

106

104

102

100

98

96

94

N D J F M A MMonth

J J A S O N

Fig. 21.9 Relationship between feeding pattern andfertility in !Kung San of Botswana. The relationshipbetween weight change and fertility in !Kung San ofBotswana. The !Kung San of Botswana were a tribe ofhunter–gatherers until about 30 years ago. The bodyweight of the men and women in this tribe varied greatlythroughout the year depending on the availability of food supplies. In the summer months, the food supplywas more abundant and the body weight increased;conversely, body weight decreased throughout the wintermonths. The number of births in the tribe peaked 9months after the achievement of peak body weight. Theseanthropological data illustrate how availability of foodcan regulate fertility patterns. (Reproduced from Van DerWalt 1978.)

Page 314: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

298 chapter 21

39 men with end-stage renal disease on hemodia-lysis who did not have diabetes mellitus, 24 (63%)had serum total and free testosterone levels belowthe lower limit of normal male range (Singh et al.2001). There is a high prevalence of sexual dysfunc-tion and spermatogenic abnormalities in men onhemodialysis (Handelsman & Dong 1993). Musclemass is decreased and fat mass increased, and muscle performance and physical function aremarkedly impaired in men receiving hemodialysis(Kopple 1999; Johansen et al. 2001). While androgendeficiency might contribute to the complex patho-physiology of sexual dysfunction and sarcopenia in men on hemodialysis, we do not know if any ofthese physiologic derangements can be reversed byandrogen replacement.

The pathophysiology of hypogonadism in chronicillness is multifactorial; defects exist at all levels ofthe hypothalamic–pituitary–testicular axis (Bross et al. 1998). Malnutrition, mediators and products of the systemic inflammatory response, drugs suchas ketoconazole and metabolic abnormalities pro-duced by the systemic illness all contribute to adecline in testosterone production.

Low testosterone levels correlate with poor disease out-come. Low testosterone levels correlate with adversedisease outcome in HIV-infected men. Serum tes-tosterone levels are lower in HIV-infected men who have lost weight than in those who have not(Coodley et al. 1994). Longitudinal follow up ofHIV-infected homosexual men reveals a progressivedecrease in serum testosterone levels; this decreaseis much greater in HIV-infected men who progressto acquired immunodeficiency syndrome (AIDS)than in those who do not. Serum testosterone levelsin HIV-infected men decline early in the course ofevents that culminate in wasting (Dobs et al. 1996).Testosterone levels correlate with muscle mass andexercise capacity in HIV-infected men (Grinspoon et al. 1996). Although, patients with HIV infectionmay lose both fat and lean tissue, the loss of leanbody mass is an important aspect of the weight lossassociated with wasting. There is a high prevalenceof sexual dysfunction in HIV-infected men. With the increasing life expectancy of HIV-infected men,frailty and sexual dysfunction have emerged asimportant quality of life issues.

Similarly, muscle mass, strength and perform-ance and physical function are markedly impairedin patients on hemodialysis ( Johansen et al. 2001).Exercise tolerance is attenuated (Kopple 1999;Johansen et al. 2001); peak oxygen uptake is typ-ically reduced to roughly half the level predicted for healthy subjects. Although the etiology of sar-copenia in end-stage renal disease is complex, thedecrease in testosterone levels, a contributor to lossof muscle mass and dysfunction, is potentiallyreversible.

Changes in testosterone levels during exercise

The literature on the effects of exercise on testicu-lar function is controversial in part because the published studies differed significantly in the type,mode, intensity and duration of exercise. Further-more, few studies controlled for the confoundinginfluence of nutritional intake and physical activitylevel. Therefore, it is not surprising that bothincreases and decreases in circulating testosteronelevels have been reported in men undergoing exer-cise training. This is in contrast to women under-going heavy endurance exercise training in whomconsistent disruptions of menstrual cycle and ovarian estrogen production have been observed(Frisch & Revelle 1970; Mahna et al. 1973; Warren1980; Baker et al. 1981; Veldhuis et al. 1985; Loucks et al. 1989; Cumming 1996). The onset of menarche is often delayed in ballet dancers and in girls who are engaged in heavy exercise (Frisch et al. 1980).Similarly, a high prevalence of menstrual irregular-ities, attenuated gonadotropin secretion and highcortisol levels has been documented in female run-ners (Baker et al. 1981; Villanueva et al. 1986); how-ever, the literature on the literature on the effects ofexercise on reproductive function in men is sparseand less lucid (MacConnie et al. 1986; Skarda &Burge 1998).

Broadly, exercise-training regimens can beclassified into endurance and resistance exerciseregimens. Although serum testosterone levels mayincrease in anticipation of agonistic events or anendurance exercise, most studies are in agreementthat mild endurance exercise has little or no clin-ically significant effect on circulating testosterone

Page 315: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 299

levels (Skarda & Burge 1998; Smilios et al. 2003). Incontrast, very severe endurance exercise training asexemplified by long distance running, especially if accompanied by significant energy drain andweight loss, is associated with the lowering of serumtestosterone levels (Remes et al. 1979; Häkkinen et al.1985; MacConnie et al. 1986; Dressendorfer & Wade1991; Skarda & Burge 1998). For instance, male dis-tance runners who ran on average 92 km weeklywere found to have 10% lower bone mineral densityat the lumbar spine than a control group of men whodid not run (Frost 1992). In general, the distance run correlates inversely with vertebral and femoralbone mineral densities; the longer the distance run,the greater the energy drain, the lower the bonemineral density. In one report of men participatingin a 15-day 400-km race, serum testosterone levelsdeclined significantly while cortisol levels increased(Dressendorfer & Wade 1991). Therefore, it is notsurprising that some of the lowest testosterone levels have been reported in army recruits who arein intense boot camp training.

Remarkably, men who run 15–20 miles weeklyhave higher bone mineral density than age-matchedcontrols who do not run (Frost 1992; Burrows et al.2003). Similarly, rowers have been reported to havea higher bone mineral density than sedentary con-trols. In another study, triathletes did not differ fromsedentary controls in their bone mineral density.Thus, mild to moderate endurance exercise trainingmay have beneficial effects on bone mineral densitywith little or no effect on testosterone concentrations;very intense endurance exercise training lowers testo-sterone concentrations and decreases bone mineraldensity (Heinonen et al. 1995; Burrows et al. 2003).

There is agreement that long distance runnersmay have significant energy deficit and nutritionaldeficiencies that may affect bone mineral densityindependent of the effects of exercise on testo-sterone levels (Burrows et al. 2003). Indeed, some

studies suggest that only nutritionally deleteriouseating behaviors are associated with lower bonemineral density and increased fracture risk. Fur-thermore, some of the deleterious effects of lowersex hormone levels on bone mineral density may beoffset by the direct beneficial effects of exercise andphysical activity on bone mineral density.

Frost (1992) has hypothesized that the effect ofexercise on bone mineral density depends cruciallyon the force applied to the limbs during exercise. He has speculated that only exercise regimens that apply forces on the limbs in excess of a certainthreshold level are capable of eliciting a boneremodeling response, whereas long distance run-ning that exerts forces of a lower magnitude (5–10 times the body weight) on the limbs does notincrease bone mineral density. According to thishypothesis, the magnitude of bone loading is moreimportant than the exercise mode or number ofcycles. Thus, lean marathon runners or boot camptrainees, with significant energy deficits, would beexpected to have lower testosterone levels, no coun-terbalancing beneficial effects of bone loading, andconsequently lower bone mineral density.

Previous studies have reported either no changeor modest increments in serum total testosteroneconcentrations during regimens of resistance exer-cise training (Remes et al. 1979; Truls et al. 2000;Ahtiainen et al. 2003). The small changes in testo-sterone concentrations reported in some studieswere not sustained after completion of the exercise;in fact some studies have reported a significantdecline in free testosterone levels during recoveryfrom exercise. SHBG concentrations generally didnot change during the course of resistance exercise.Some studies have reported increases in the testo-sterone to cortisol ratio during progressive trainingfor maximal strength gains. There is considerableinterindividual variability in hormonal response toresistance exercise training.

References

Ahtiainen, J.P., Pakarinen, A., Kraemer,W.J. & Häkkinen, K. (2003) Acutehormonal and neuromuscular responses and recovery to forced vs.maximum repetitions multipleresistance exercises. International

Journal of Sports Medicine 24(6), 410–418.

Apter, D., Pakarmen, A. & Vihko, R. (1978)Serum prolactin, FSH and LH duringpuberty in girls and boys. ActaPaediatrica Scandinavica 67, 417–423.

Apter, D., Cacciatore, B., Alfthan, H. &Stenmain, U.-H. (1989) Serum LHconcentrations increase 100-fold infemales from 7 years of age toadulthood, as measured by time-resolved immunofluorometric assay.

Page 316: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

300 chapter 21

Journal of Clinical Endocrinology andMetabolism 68, 53–57.

Arver, S.A., Sinha-Hikim, I., Beall, G. et al. (1999) Dihdrotestosterone andtestosterone levels in HIV-infected men.Journal of Andrology 20, 611–618.

Aubert, M.L., Pierroz, D.D., Gruaz, N.M. et al. (1998) Metabolic control of sexual function and growth: role ofneuropeptide Y and leptin. Molecular and Cellular Endocrinology 140, 107–113.

Badger, T.M., Edcox, C.E., Meyer, E.R.,Bell, R.D. & Cicero, T.J. (1978)Simultaneous changes in tissue andserum levels of LH, FSH and LH/FSHreleasing factor after castration in themale rat. Endocrinology 102, 136–141.

Baenziger, J.U. (1990) The asparagine-linked ohgosaccharides of theglycoprotein hormones. In: GlycoproteinHormones (Chin, W.W. & Boime, L.,eds.). Serono Symposia, Norwell, MA:11–18.

Baker, E.R., Mathur, R.S., Kirk, R.S. &Williamson, H.O. (1981) Female runnersand secondary amenorrhea: correlationwith age, parity, mileage, and plasmahormonal and sex-hormone buildingconcentrations. Fertility and Sterility 36,183–189.

Bardin, C.W., Ross, G.T., Rifkind, A.B.,Cargille, C.M. & Lipsett, M.B. (1969)Studies of the pituitary Leydig cell axisin young men with hypogonadotropichypogonadism and hyposmia.Comparison with normal men,prepubertal boys and hypopituitarypatients. Journal of Clinical Investigation48, 2046–2056.

Bates, G.W., Bates, S.R. & Whitworth, N.S.(1982) Reproductive failure in womenwho practice weight control. Fertility and Sterility 37, 373–378.

Belchetz, P.E., Plant, T.M., Naka, Y.,Keogh, E.J. & Knobil, E. (1978)Hypophyseal responses to continuousand intermittent delivery ofhypothalamic GnRH. Science 202,631–643.

Bhasin, S., Fielder, T.J. & Swerdloff, R.S.(1987) Testosterone selectively increasesserum FSH but not LH in GnRHantagonist treated male rats: evidencefor differential regulation of LH andFSH secretion. Biology of Reproduction 37,55–59.

Boccabella, A.V. (1963) Reinitiation and restoration of spermatogenesis with testosterone propionate and other hormones after a long termposthypophysectomy regression period.Endocrinology 72, 787–798.

Clarke, I.J. & Cummins, J.T. (1999) Direct pituitary effects of estrogen and progesterone on gonadotropinsecretion in the ovariectomized ewe.Neuroendocrinology 39, 267–274.

Clarke, I.J. & Henry, B.A. (1999) Leptin andreproduction. Biology of Reproduction 4,48–55.

Clements, J.A., Reves, F.L., Winter, J.S.D. & Faiman, C. (1980) Ontogenesis ofGnRH in the human fetal hypothalamus.Proceedings of the Society for ExperimentalBiology and Medicine 163, 437–444.

Cole, L.A., Perini, F., Birken, S. & Ruddon,R. (1984) An oligosacchande of the O-linked type distinguishes the free fromthe combined form of hCG α subunit.Biochemical and Biophysical ResearchCommunications 122, 1260–1267.

Conn, P.M., Chafouleas, J.C., Rogers, D.C.& Means, A.R. (1981) Gonadotropinreleasing hormone stimulatescalmodulin redistribution in ratpituitary. Nature 292, 264–265.

Conn, P.M., Rogers, D.C. & McNeil, R.(1982) Potency enhancement ofgonadotropin-releasing hormoneagonist GnRH-mediatedmicroaggregation stimulatesgonadotropin release. Endocrinology 111,335–337.

Coodley, G.O., Loveless, M.O., Nelson,H.D. & Coodley, M.K. (1994). Endocrinefunction in HIV wasting syndrome.Journal of Acquired Immune DeficiencySyndromes and Human Retrovirology 7,46–51.

Craig, B.W., Brown, R. & Everhart, J. (1989)Effects of progressive resistance trainingon growth hormone and testosteronelevels in young and elderly subjects.Mechanisms of Ageing and Development49(2), 159–169.

Crowley, W.F., Jr., Whitcomb, R.W.,Jameson, J.L. et al. (1991) Theneuroendocrine control of humanreproduction in the male. Recent Progressin Hormone Research 47, 27–62.

Culler, M.D. & Negro-Vilar, A. (1990)Destruction of testicular Leydig cellsreveals a role of endogenous inhibin inregulating follicle-stimulating hormonesecretion in the adult male rat. Molecularand Cellular Endocrinology 70, 89–98.

Cumming, D. (1996) Exercise-associatedamenorrhea, low bone mineral densityand oestradiol replacement therapy.Archives of Internal Medicine 156,2193–2195.

Cunningham, M.J., Clifton, D.K. & Steiner,R.A. (1999) Leptin’s actions on thereproductive axis: perspectives and

Brinkmann, A.O. (2001) Molecular basis of androgen insensitivity. Molecular andCellular Endocrinology 179(1–2), 105–109.

Bross, R., Casaburi, R., Storer, T.W. &Bhasin, S. (1998) Androgen effects onbody composition and muscle function:implications for the use of androgens asanabolic agents in sarcopenic states.Bailliere’s Clinical Endocrinology andMetabolism 12(3), 365–378.

Bross, R., Singh, A.B. & Bhasin, S. (2000) In: Atlas of Clinical Endocrinology: HumanNutrition and Obesity (Heber, D., ed.).Blackwell Science, Philadelphia, PA.

Burger, H.G. & Igarashi, M. (1988) Inhibin:definition and nomenclature, includingrelated substances. Journal of ClinicalEndocrinology and Metabolism 66,885–886.

Burrows, M., Nevill, A.M., Bird, S. &Simpson, D. (2003) Physiological factorsassociated with low bone mineraldensity in female endurance runners.British Journal of Sports Medicine 37(1),67–71.

Cai, L.Q., Zhu, Y.S., Katz, M.D. et al. (1996)5α-reductase-2 gene mutations in theDominican Republic. Journal of ClinicalEndocrinology and Metabolism 81(5),1730–1735.

Carani, C., Qin, K., Simoni, M. et al. (1997)Effect of testosterone and estradiol in aman with aromatase deficiency. NewEngland Journal of Medicine 337(2), 91–95.

Casaburi, R., Goren, S. & Bhasin, S. (1996)Substantial prevalence of low anabolichormone levels in COPD patientsundergoing rehabilitation. AmericanJournal of Respiratory and Critical CareMedicine 153, A128.

Casella, R., Maduro, M.R., Lipshultz, L.I. & Lamb, D.J. (2001) Significance of thepolyglutamine tract polymorphism inthe androgen receptor. Urology 58(5),651–656.

Childs, G.V., Ellison, D.G., Foster, L. & Ramaley, J.A. (1981) Postnatalmaturation of gonadotropes in the malerat pituitary. Endocrinology 109,1683–1693.

Childs, G.V., Llovd, J.M., Unabia, G. et al.(1987) Detection of LHβ mRNA inindividual gonadotropes aftercastration: use of a new in situhybridization method with a photobiotinylated complementary RNAprobe. Molecular Endocrinology(Baltimore, Md) 1, 926–932.

Clark, B.J. & Stocco, D.M. (1982) STAR. A tissue specific acute mediator ofsteroidogenesis. Trends in Endocrinologyand Metabolism 91, R1–R6.

Page 317: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 301

mechanisms. Biology of Reproduction 60,216–222.

Dai, W.S., Kuller, L.H., LaPorte, R.E. et al.(1981) The epidemiology of plasmatestosterone levels in middle-aged men.American Journal of Epidemiology 114,804–816.

Dobs, A.S., Few, W.L., Blackman, M.R. et al. (1996) Serum hormones in menwith human immunodeficiency virus-associated wasting. Journal of ClinicalEndocrinology and Metabolism 81,4108–4112.

Dressendorfer, R.H. & Wade, C.E. (1991)Effects of a 15-d race on plasma steroidlevels and leg muscle fitness in runners.Medicine and Science in Sports and Exercise23(8), 954–958.

Drinkwater, B.L., Nilson, K., Chestnut,C.H. et al. (1984) Bone mineral content ofamenorrheic and eumenorrheic women.New England Journal of Medicine 311,2777–2782.

Dufau, M.L. (1988) Endocrine regulationand communicating functions of theLeydig cell. Annual Review of Physiology50, 483–508.

Feldman, H.A., Longcope, C., Derby, C.A.et al. (2002) Age trends in the level ofserum testosterone and other hormonesin middle-aged men: longitudinalresults from the Massachusetts maleaging study. Journal of ClinicalEndocrinology and Metabolism 87,589–598.

Ferrini, R.L. & Barrett-Connor, E. (1998)Sex hormones and age: a cross-sectionalstudy of testosterone and estradiol andtheir bioavailable fractions incommunity-dwelling men. AmericanJournal of Epidemiology 147, 750–754.

Fiddes, J.C. & Talmadge, K. (1984)Structure, expression and evolution ofthe genes for the human glycoproteinhormones. Recent Progress in HormoneResearch 40, 43–78.

Finkel, D.M., Phillips, J.L. & Snyder, P.J.(1985) Stimulation of spermatogenesisby gonadotropins in men withhypogonadotropic hypogonadism. NewEngland Journal of Medicine 313, 651–655.

Finkelstein, J.S., O’Dea, St. L, Whitcomb,R.W. & Crowley, W.E., Jr. (1991a) Sexsteroid control of gondotropin secretionin the human male II. Effects of estradioladministration in normal and GnRHdeficient men. Journal of ClinicalEndocrinology and Metabolism 73,621–628.

Finkelstein, J.S., Whitcomb, R.W., O’Dea,St. L. et al. (1991b) Sex steroid control of gonadotropin secretion in the

in hypogondal men with acquiredimmunodeficiency syndrome andwasting. Journal of Clinical Endocrinologyand Metabolism 81, 4051–4058.

Grossman, M., Weintraub, B.D. &Szkudlinski, M.W. (1997) Novel insightsinto the molecular mechanisms ofhuman thyrotropin action: structural,physiological, and therapeuticimplications for the glycoproteinhormone family. Endocrine Reviews 18,476–501.

Grumbach, M.M. & Auchus, R.J. (1999)Estrogen: consequences andimplications of human mutations insynthesis and action. Journal of ClinicalEndocrinology and Metabolism 84(12),4677–4694.

Häkkinen, K., Pakarinen, A., Alen, M. & Komi, P.V. (1985) Serum hormonesduring prolonged training ofneuromuscular performance. EuropeanJournal of Applied Physiology andOccupational Physiology 53(4), 287–293.

Haisenleder, D.J., Katt, J.A., Ortolano, G.A.et al. (1988) Influence of GnRH pulseamplitude, frequency and treatmentduration on the regulation of LH subunitmRNAs and LH secretion. MolecularEndocrinology (Baltimore, Md) 12,338–343.

Haisenleder, D.J., Dalkin, A.C., Ortolano,G.X., Marshall, J.C. & Shupnik, M.A.(1991) A pulsatile GnRH stimulus isrequired to increase transcription of the gonadotropin subunit genes:evidence for differential regulation oftranscription by pulse frequency in vivo.Endocrinology 128, 509–517.

Hall, S.H. & Miller, W.L. (1986) Regulationof ovine pituitary glycoprotein hormoneα subunit mRNA by 17-β-estradiol incell culture. Biology of Reproduction 34,533–542.

Handelsman, D.J. & Dong, Q. (1993)Hypothalamic–pituitary–gonadal axis inchronic renal failure. Endocrinology andMetabolism Clinics of North America 22,145–161.

Harman, S.M., Tsitouras, P.D., Cost, P.T. & Blackman, M.R. (1982) Reproductivehormones in aging men II. Basalpituitary gonadotropins andgonadotropin responses to LHRH.Journal of Clinical Endocrinology andMetabolism 54, 547–551.

Harman, S.M., Metter, E.J., Tobin, J.D.,Pearson, J. & Blackman, M.R. (2001)Longitudinal effects of aging on serumtotal and free testosterone levels inhealthy men. Baltimore LongitudinalStudy of Aging. Journal of Clinical

human male I. Effects of testosteroneadministration in normal and GnRHdeficient men. Journal of ClinicalEndocrinology and Metabolism 73,609–620.

Foster, D.L. & Nagatani, S. (1999)Physiological perspectives on leptin as aregulator of reproduction: role in timingpuberty. Biology of Reproduction 60,205–215.

Frisch, R.E. (1989) Body weight andreproduction. Science 246, 432–436.

Frisch, R.E. & McArthur, J.W. (1974)Menstrual cycles: fatness as adeterminant of minimum weight forheight necessary for their maintenanceor onset. Science 185, 949–995.

Frisch, R.E. & Revelle, R.L. (1970) Heightand weight at menarche and ahypothesis of critical body weights andadolescent events. Science 169, 397–398.

Frisch, R.E., Wushak, G. & Vincent, L.(1980) Delayed menarche andamenorrhea of ballet dancers. NewEngland Journal of Medicine 303, 17–19.

Frost, H.M. (1992) The role of changes inmechanical usage set points in thepathogenesis of osteoporosis. Journal ofBone and Mineral Research 7, 253–261.

Gharib, S.D., Bowers, S.M., Need, L.R. &Chin, W.W. (1986) Regulation of rat LHsubunit mRNAs by gonadal steroidhormones. Journal of Clinical Investigation77, 582–589.

Gharib, S.D., Wierman, M.E., Badger, T.M.& Chin, W.W. (1987) Sex steroidhormone regulation of folicle-stimulating hormone subunit messengerribonucleic acid (mRNA) by gonadalsteroid hormones. Journal of ClinicalInvestigation 80, 294–299.

Gharib, S.D., Wierman, M.E., Shupnik,M.A. & Chin, W.W. (1990) Molecularbiology of the pituitary gonadotropins.Endocrine Reviews 11, 177–199.

Glass, A.R. & Denster, P.A. (1987)Amenorrhea in Olympic marathonrunners. Fertility and Sterility 4, 740–745.

Gormley, G. & Rittmaster, R. (1992) Effectof finasteride, a 5-α reductase inhibitor,on serum gonadotropins in normal men.Journal of Clinical Endocrinology andMetabolism 75, 484–488.

Gray, A., Berlin, J.A., McKinlay, J.B. &Longcope, C. (1991) An examination ofresearch design effects on the associationof testosterone and male aging: results of a meta-analysis. Journal of ClinicalEpidemiology 44, 671–684.

Grinspoon, S., Corcoran, C., Lee, K. et al.(1996) Loss of lean body and musclemass correlates with androgen levels

Page 318: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

302 chapter 21

Endocrinology and Metabolism 86,724–731.

Heinonen, A., Oja, P., Kannus, P. et al.(1995) Bone mineral density in femaleathletes representing sports withdifferent loading characteristics of theskeleton. Bone 17, 197–203.

Horton, R. (1978) Sex steroid productionand secretion in the male. Andrologia10(3), 183–194.

Huhtaniemi, I. & Toppari, J. (1995)Endocrine, paracrine and autocrineregulation of testicular steroidogenesis.Advances in Experimental Medicine andBiology 377, 33–54.

Husmann, D.A. (1991) Inhibition oftesticular descent in the rat by antiandrogens. Endocrinology 73, 1162A.

Ingraham, H.A., Chen, R.P., Mangalam,H.J. et al. (1988) A tissue-specifictranscription factor containing ahomeodomain specifies a pituitaryphenotype. Cell 55, 519–529.

Jakachi, R.I., Kelch, R.P., Sander, S.E. et al.(1982) Pulsatile release of LH in children.Journal of Clinical Endocrinology andMetabolism 55, 453–458.

Johansen, K.L., Chertow, G.M., da Silva,M., Carey, S. & Painter, P. (2001)Determinants of physical performancein ambulatory patients on hemodialysis.Kidney International 60(4), 1586–1591.

Jones, M.E., Thorburn, A.W., Britt, K.L. et al. (2000) Aromatase-deficient (ArKO)mice have a phenotype of increasedadiposity. Proceedings of the NationalAcademy of Sciences of the United States ofAmerica 97(23), 12 735–12 740.

Kaufman, J.M. & Vermeulen, A. (1997)Declining gonadal function in elderlymen. Bailliere’s Clinical Endocrinology and Metabolism 11, 289–309.

Keman, D., Madigan, M.B., Bardin, W.C. & Chen, L.-C.C. (1989) Expression andregulation of testicular inhibin α subunitgene in vivo and in vitro. MolecularEndocrinology (Baltimore, Md) 3, 29–35.

Keys, A., Brozek, J., Henschel, A. et al.(1950) The Biology of Human Starvation.University of Minnesota Press,Minneapolis.

Kim, W.H., Swerdloff, R.S. & Bhasin, S.(1988a) Regulation of α and rat LH βmRNAs during GnRH agonist treatmentin vivo in the male rat. Endocrinology 123,2111–2116.

Kim, W.H., Yuan, Q.X., Swerdloff, R.S. &Bhasin, S. (1988b) Regulation of α andLH β mRNAs during stimulatory anddownregulatory phases of GnRH action. Biology of Reproduction 39,847–853.

Ling, N., Ying, S.-Y., Ueno, N. et al. (1985)Isolation and partial characterization ofa Mr 32000 protein with inhibin activityfrom porcine follicular fluid. Proceedingsof the National Academy of Sciences of theUnited States of America 182, 7217–7221.

Longcope, C. (1973) The effect of humanchorionic gonadotropin on plasmasteroid levels in young and old men.Steroids 21, 583–592.

Longcope, C. & Fineberg, S.E. (1985)Production and metabolism ofdihydrotestosterone in peripheraltissues. Journal of Steroid Biochemistry23(4), 415–419.

Loucks, A.B., Mortola, J.F., Girton, L. &Yen, S.S.C. (1989) Alterations in thehypothalamic–pituitary–ovarian and thehypothalamic–pituitary–adrenal axis in athletic women. Journal of ClinicalEndocrinology and Metabolism 68,402–411.

McCann, S.M., Kimura, M., Walczewska,A. et al. (1998) Hypothalamic control ofFSH and LH by FSH-EF, LHRH,cytokines, leptin and nitric oxide.Neuroimmunomodulation 5, 193–202.

MacConnie, S.E., Barkan, A., Lampman,R.M. et al. (1986) Decreasedhypothalamic gonadotropin-releasinghoromone secretion in male marathonrunners. New England Journal of Medicine315, 411–417.

McCullagh, D.R. (1932) Dual endocrineactivity of the testes. Science 76, 19–23.

McFarland, K.C., Sprengel, R., Phillip, H. et al. (1989) Lutropin/choriogonadotropin receptor: anunusual member of the G-proteincoupled receptor family. Science 245,494–499.

McLachlan, R.I., Matsumoto, A.M., Burger, H.G., de Kretser, D.M. &Bremmer, W.J. (1988) Relative roles of follicle-stimulating hormone andluteinizing hormone in the control ofinhibin secretion in normal men. Journalof Clinical Investigation 82, 880–884.

McPhaul, M.J., Marcelli, M., Zoppi, S.,Griffin, J.E. & Wilson, J.D. (1993) Geneticbasis of endocrine disease. 4. Thespectrum of mutations in the androgenreceptor gene that causes androgenresistance. Journal of ClinicalEndocrinology and Metabolism 76(1),17–23.

Mahna, R.M., Harper, A.B., Avent, H.H. &Campbell, D.E. (1973) Age at menarchein athletes and non-athletes. Medicineand Science in Sports 5, 11–13.

Marshall, G.R., Wickings, E.J., Ludecke,D.K. & Nieschlag, E. (1983) Stimulation

Knobil, E. (1980) The neuroendocrinecontrol of the menstrual cycle. RecentProgress in Hormone Research 36, 53–88.

Kopple, J.D. (1999) Pathophysiology ofprotein-energy wasting in chronic renal failure. Journal of Nutrition129(1S suppl.), 247S–251S.

Kornfeld, R. & Kornfeld, S. (1976)Comparative aspects of glycoproteinstructure. Annual Review of Biochemisty45, 217–237.

Kovacs, K. & Horvath, E. (1985)Morphology of adenohyphyseal cellsand, pituitary adenomas. In: ThePituitary Gland (Martini, L., eds.). Raven Press, New York: 25–56.

Krummen, L.A., Toppari, J., Kim, W.H. et al. (1989) Regulation of testicularinhibin subunit messenger ribonucleicacid levels in vivo; effects ofhypophysectomy and selective follicle-stimulating hormone replacement.Endocrinology 125, 1630–1637.

Krummen, L.A., Morelos, B.S. & Bhasin, S.(1990) The role of LH in regulation oftesticular inhibin α and βB subunitmRNAs in immature and adult animals.Endocrinology 127, 1097–1104.

Larrea, F., Carino, C., Hardy, D.O., Musto,N.A. & Catterall, J.F. (1995) Geneticvariations in human testosterone–estradiol binding globulin. Journal ofSteroid Biochemistry and Molecular Biology53(1–6), 553–559.

Layman, L.C. (1999) Mutations in humangonadotropin genes and theirphysiologic significance in puberty andreproduction. Fertility and Sterility 71(2),201–218.

Lee, D.K. & Chang, C. (2003) Endocrinemechanisms of disease: expression anddegradation of androgen receptor:mechanism and clinical implication.Journal of Clinical Endocrinology andMetabolism 88(9), 4043–4054.

Lee, P.A. (1988) Pubertal neuroendocrinematuration: early differentiation andstages of development. Adolescent andPediatric Gynecology 1, 3–14.

Lee, P.A., Yenakis, T., Winer, J. &Matsenbaugh, S. (1976) Puberty in girls: correlation of serum levels ofgonadotropins, prolactin, androgens,estrogens and progestins with physicalchanges. Journal of Clinical Endocrinologyand Metabolism 43, 775–784.

Legouis, R., Hardelin, J.P., Levilliers, J. et al. (1991) The candidate gene for the X-linked Kallmann syndrome encodes aprotein related to adhesion molecules.Cell 67(2), 423–435.

Page 319: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 303

of spermatogenesis in stalk-sectionalrhesus monkeys by testosterone alone.Journal of Clinical Endocrinology andMetabolism 57, 152–159.

Mason, A.J., Hayflick, J.S., Ling, N. et al.(1985) Complementary DNA sequencesof ovarian follicular fluid inhibin showprecursor structure and homology withtransforming growth factor-β. Nature318, 659–663.

Mather, J.P., Attie, K.M., Woodruff, T.K.,Rice, G.C. & Phillips, D.M. (1990)Activin stimulates spermatogonialproliferation in germ–Sertoli cell co-cultures from immature rat testis.Endocrinology 127, 3206–3214.

Matsumoto, A.M. (2002) Andropause:clinical implications of the decline inserum testosterone levels with aging inmen. Journals of Gerontology. Series A,Biological Sciences and Medical Sciences57(2), M76–M99.

Matsumoto, A.M. & Bremner, W.J. (1984)Modulation of pulsatile gonadotropinsecretion by testosterone in men. Journalof Clinical Endocrinology and Metabolism58, 609–614.

Matsumoto, A.M., Karpas, A.E., Paulsen, C.A. & Bremner, W.J. (1983)Reinitiation of sperm production ingonadotropin-suppressed normal menby administration of FSH. Journal ofClinical Investigation 72, 1005–1015.

Matsumoto, A.M., Paulsen, C.A. &Bremner, W.J. (1984) Stimulation ofsperm production by human LH ingonadotropin suppressed normal men.Journal of Clinical Endocrinology andMetabolism 59, 882–887.

Mendonca, B.B., Inacio, M., Costa, E.M. etal. (1996) Male pseudohermaphroditismdue to steroid 5 α-reductase 2 deficiency.Diagnosis, psychological evaluation,and management. Medicine; AnalyticalReviews of General Medicine, Neurology,Psychiatry, Dermatology, and Pediatrics75(2), 64–76.

Mercer, J.E., Clements, J.E., Funder, J.W. & Clarke, I.J. (1988) LH β mRNA levelsare regulated primarily by GnRH andnot by negative estrogen feedback on thepituitary. Neuroendocrinology 47, 63–66.

Mohamed-Ali, V., Pinkney, J.H. &Coppack, S.W. (1998) Adipose tissue asan endocrine and paracrine organ.International Journal of Obesity and RelatedMetabolic Disorders 22, 1145–1158.

Mori, M. & Marsh, J.M. (1982) The site ofLH stimulation of steroidogenesis inmitochondria of rat corpus luteum.Journal of Biological Chemistry 257,6178–6183.

Rietschel, P., Corcoran, C., Stanley, T. et al.(2000) Prevalence of hypogonadismamong men with weight loss related to human immunodeficiency virusinfection who were receiving highlyactive antiretroviral therapy. ClinicalInfectious Diseases 31(5), 1240–1244.

Rivier, C., Rivier, J. & Vale, W. (1986)Inhibin mediated feedback control ofFSH secretion in the female rat. Science234, 205–208.

Rock, C.L., Gorenflo, D.W., Drewnowski,A. & Demitrack, M.A. (1996) Nutritionalcharacteristics, eating pathology, andhormonal status in young women.American Journal of Clinical Nutrition 64,566–571.

Rommerts, F.F.G. (2004) Testosterone: an overview of biosynthesis transportmetabolism and non-genomic actions.In: Testosterone: Action, Deficiency andSubstitution (Nieschlag, E. & Behr, H.M.,eds). Cambridge University Press, New York: 1–37.

Rosner, W. (1991) Plasma steroid-bindingproteins. Endocrinology and MetabolismClinics of North America 20(4), 697–720.

Russell, D.W. & Wilson, J.D. (1994) Steroid5-α reductase: two gene/two enzymes.Annual Review of Biochemistry 63, 25–61.

Ryan, R.J., Keutmann, H.T., Charlesworth,M.C. et al. (1987) Structure functionrelationships of gonadotropins. RecentProgress in Hormone Research 43, 383–429.

Saade, G., London, D.R. & Clayton, R.N.(1989) The interaction of GnRH andestradiol on LH and prolactin geneexpression in hypogonadal (hpg) mice.Endocrinology 124, 1744–1753.

Sairam, M.R. (1983) Gonadotropichormones: relationship betweenstructure and function with emphasis on antagonists. In: Hormonal Proteins andPeptides, vol. 11 (Li, C.H., ed.). AcademicPress, New York: 1–79.

Santen, R.J. (1975) Is aromatization oftestosterone to estradiol required forinhibition of LH secretion in men?Journal of Clinical Investigation 56,1555–1563.

Scheckter, C.B., Matsumoto, A.M. &Bremner, W.J. (1989) Testosteroneadministration inhibits gonadotropinsecretion by an effect directly at thehuman pituitary. Journal of ClinicalEndocrinology and Metabolism 68,397–401.

Schwanzel-Fukuda, M. & Pfaff, D.W.(1989) Origin of luteinizing hormonereleasing hormone neurons. Nature 338,161–163.

Moriarty, G.C. (1973) Adenohypophysis:ultrastructural cytochemistry. A review.Journal of Histochemistry andCytochemistry 21, 855–892.

Morley, J.E., Kaiser, F.E., Perry, H.M., 3rdet al. (1997) Longitudinal changes intestosterone, LH, and FSH in healthy,older men. Metabolism 46, 410–413.

Najmabadi, H., Rosenberg, L.A., Yuan,Q.X. et al. (1993) Transcriptional andpost-transcriptional regulation ofintribin α subunit gene expression in ratsertoli cell by 8-bromo cAMP. MolecularEndocrinology (Baltimore, Md) 7, 469–476.

Neill, J.D., Patton, J.M., Daily, R.A., Tsour,R.C. & Tindall, G.T. (1977) LHRH inpituitary stalk brood of rhesus monkeys:relationship to level of LH release.Endocrinology 101, 430–434.

Pardridge, W.M. (1987) Plasma protein-mediated transport of steroid andthyroid hormones. American Journal ofPhysiology 252(2 Pt. 1), E157–E164.

Parks, J.S., Brown, M.R., Hurley, D.L.,Phelps, C.J. & Wajnrajch, M.P. (1999)Heritable disorders of pituitarydevelopment. Journal of ClinicalEndocrinology and Metabolism 84,4362–4370.

Penny, R., Goldstein, I.P. & Frasier, S.D.(1978) Gonadotropin excretion and body composition. Pediatrics 61,294–300.

Pincus, S.M., Veldhuis, J.D., Mulligan, T.,Iranmanesh, A. & Evans, W.S. (1997)Effects of age on the irregularity of LHand FSH serum concentrations inwomen and men. American Journal ofPhysiology 273(5 Pt 1), E989–E995.

Pirke, K.M. & Doerr, P. (1973) Age relatedchanges and interrelationships betweenplasma testosterone, oestradiol andtestosterone-binding globulin in normaladult males. Acta Endocrinologica 74,792–800.

Prince, F.P. (2001) The triphasic nature ofLeydig cell development in humans, andcomments on nomenclature. Journal ofEndocrinology 168(2), 213–216.

Rebar, R., Judd, H.L., Yen, S.S.C. et al.(1976) Characterization of theinappropriate gonadotropin secretion inpolyvistic ovary syndrome. Journal ofClinical Investigation 57, 1320–1325.

Remes, K., Kuoppasalmi, K. &Adlercreutz, H. (1979) Effect of long-term physical training on plasmatestosterone, androstenedione,luteinizing hormone and sex-hormone-binding globulin capacity. ScandinavianJournal of Clinical and LaboratoryInvestigation 39(8), 743–749.

Page 320: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

304 chapter 21

Schwartz, M.W., Baskin, D.G., Kaiyala, K.J.& Woods, S.C. (1999) Model for theregulation of energy balance andadiposity by the central nervous system.American Journal of Clinical Nutrition 69,584–596.

Scully, K.M. & Rosenfeld, M.G. (2002)Pituitary development: regulatory codesin mammalian organogenesis. Science295(5563), 2231–2235.

Seth, A., Stanley, S., Jethwa, P. et al. (2004)Galanin-like peptide stimulates therelease of gonadotropin-releasinghormone in vitro and may mediate theeffects of leptin on the hypothalamo–pituitary–gonadal axis. Endocrinology145(2), 743–750.

Sharpe, R.M. (1987) Testosterone and spermatogenesis. Journal ofEndocrinology 113, 1–2.

Sharpe, R.M., Donachie, K. & Cooper, L.(1988) Reevaluation of intratesticularlevel of testosterone required forquantitative maintenance ofspermatogenesis in the rat. Journal ofEndocrinology 117, 19–26.

Shupnik, M.A. (1990) Effects ofgonadotropin-releasing hormone on ratgonadotropin gene transcription in vitro:requirement for pulsatile administrationfor luteinizing hormone-β genestimulation. Molecular Endocrinology(Baltimore, Md) 4(10), 1444–1450.

Shupnik, M.A., Gharib, S.D. & Chin, W.W.(1989) Divergent effects of estradiol ongonadotropin gene transcription inpituitary fragments. MolecularEndocrinology (Baltimore, Md) 3, 474–478.

Simon, D., Preziosi, P., Barrett-Connor, E.et al. (1992) The influence of aging onplasma sex hormones in men: theTelecom Study. American Journal ofEpidemiology 135, 783–791.

Simpson, E.R. (1979) Cholesterol side chaincleavage, cytochrome P-450, and thecontrol of steroidogenesis. Molecular and Cellular Endocrinology 13, 213–227.

Singh, A.B., Norris, K., Modi, N. et al.(2001) Pharmacokinetics of atransdermal testosterone system in menwith end stage renal disease receivingmaintenance hemodialysis and healthyhypogonadal men. Journal of ClinicalEndocrinology and Metabolism 86(6),2437–2445.

Sizonenko, P.C. (1979) Endocrinology inpreadolescents and adolescents 1.Hormonal changes during normalpuberty. American Journal of Diseases ofChildren 132, 704–712.

Skarda, S.T. & Burge, M.R. (1998)Prospective evaluation of risk factors for

European Journal of Applied Physiology 82,121–128.

Ueno, N., Ling, N., Yng, S.Y. et al. (1987)Isolation and partial characterization offollistatin, a novel Mr 35000 monomericprotein which inhibits the release ofFSH. Proceedings of the National Academyof Sciences of the United States of America84, 8282–8286.

Urban, R.J., Evans, W.S., Rogol, A.D. et al.(1988) Contemporary aspects of discretepeak detection algorithms 1. Theparadigm of the LH pulse signal in men.Endocrine Reviews 9, 3–37.

Vale, W., Rivier, C., Hsueh, A.J.W. et al.(1988) Chemical and biologicalcharacterization of the inhibin family of protein hormones. Recent Progress inHormone Research 44, 1–34.

Van Der Spruy, Z.M. (1985) Nutrition andreproduction. Clinics in Obstetrics andGynaecology 12, 579–604.

Van Der Walt, L.A., Wilmsen, E.N. &Jenkins, T. (1978) Unusual sex hormonepatterns among desert-dwelling hunter gatherers. Journal of ClinicalEndocrinology and Metabolism 46,658–663.

Veldhuis, J.D., Rogol, A.D., Samojlik, E. & Ertel, N.H. (1984) Role of endogenousopiates in the expression of negativefeedback actions of androgen andestrogen on pulsatile properties of LHsecretion in man. Journal of ClinicalInvestigation 74, 47–55.

Veldhuis, J.D., Evans, W.S., Demers, L.M.et al. (1985) Altered neuroendocrineregulation of gonadotropin secretion in women distance runners. Journal ofClinical Endocrinology and Metabolism 61,557–563.

Vermeulen, A. (1988) Physiology of thetestosterone-binding globulin in man.Annals of the New York Academy ofSciences 538, 103–111.

Villanueva, A.L., Schlosser, C., Hopper, B.et al. (1986) Increased cortisol productionin women runners. Journal of ClinicalEndocrinology and Metabolism 63, 133–136.

Warren, M.P. (1980) The effects of exercise on pubertal progression andreproductive functions in girls. Journal ofClinical Endocrinology and Metabolism 51,1150–1157.

Weiss, J., Crowley, W.F., Jr. & Jameson, J.L.(1992) Pulsatile gonadotropin-releasinghormone modifies polyadenylation ofgonadotropin subunit messengerribonucleic acids. Endocrinology 130,415–420.

Whitcomb, R.W., O’Dea, St. L., Finkelstein,J.S., Heavern, D.M. & Crowley, W.F., Jr.

exercise-induced hypogonadism in malerunners. Western Journal of Medicine 169,9–12.

Smilios, I., Pilianidis, T., Karamouzis, M. & Tokmakidis, S.P. (2003) Hormonalresponses after various resistanceexercise protocols. Medicine and Sciencein Sports and Exercise 35(4), 644–654.

Smith, E.P., Boyd, J., Frank, G.R. et al.(1994) Estrogen resistance caused by amutation in the estrogen receptor genein a man. New England Journal of Medicine331, 1056–1061.

Spratt, D.I., Chin, W.W., Ridgeway, E.C. &Crowley, W.F., Jr. (1986) Administrationof low dose pulsatile GnRHadministration to GnRH deficient menregulates free α subunit secretion.Journal of Clinical Endocrinology andMetabolism 62, 102–108.

Sprengel, R., Braun, T., Nikofics, K.,Segaloff, D.L. & Seeburg, P.H. (1990) The testicular receptor for FSH: structureand functional expression of clonedcDNA. Molecular Endocrinology(Baltimore, Md) 4, 525–530.

Stager, K., Behr, R., Kleiner, I., Weinbauer,G.F., Bergmann, M. (2004) Expression of activator of CREM in the testis (ALP) during normal and impairedspermatosis genesis: correlation withCREM expression. Molecular HumanReproduction 10, 129–135.

Stein, Z., Susser, M., Saenger, G. & Marolla, F. (1973) Famine and HumanDevelopment: The Dutch Hunger Winter of 1944–1945. Oxford University Press,New York.

Steinberger, E. (1971) Hormonal control of mammalian spermatogenesis.Physiological Reviews 51, 1–22.

Steinberger, E. & Chowdhury, M. (1977)The effects of testosterone propionateand estradiol benzoate on the in vitrosynthesis of FSH. Biology of Reproduction16, 403–408.

Swerdloff, R.S., Campfield, L.A., Paiacios,A. & McClure, R.D. (1979) Suppressionof human spermatogenesis by depotandrogen: potential for malecontraception. Journal of SteroidBiochemistry 11, 663–670.

Tenover, J.S., Matsumoto, A.M., Plymate,S.R. & Bremner, W.J. (1987) The effects of aging in normal men on bioavailabletestosterone and luteinizing hormonesecretion: response to clomiphenecitrate. Journal of Clinical Endocrinologyand Metabolism 65, 1118–1126.

Truls, R., Trine, B. & Jostein, H. (2000)Hormonal responses to high- andmoderate-intensity strength exercise.

Page 321: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

regulation of testicular function 305

(1990) Utility of free α subunit as analternative neuroendocrine marker ofGnRH stimulation of the gonadotroph inthe human; evidence from normal andGnRH-deficient men. Journal of ClinicalEndocrinology and Metabolism 70,1654–1661.

Wierman, M.E. & Wang, C. (1990)Androgen selectively stimulates FSH βmRNA levels after GnRH antagonistadministration. Biology of Reproduction42, 563–571.

Wierman, M.E., Rivier, J.E. & Wang, C.(1989) GnRH dependent regulation ofgonadotropin subunit mRNA levels inthe rat. Endocrinology 124, 272–278.

Wilson, J.D., Griffin, J.E. & Russell, D.W.(1993) Steroid 5 α-reductase 2 deficiency.Endocrine Reviews 14(5), 577–593.

Winter, J.S.D., Faiman, C., Hobson, W. et al. (1975) Pituitary–gonadal relations in infancy 1. Patterns of serumgonadotropin levels from birth to 4 yearsof age in man and chimpanzee. Journal ofClinical Endocrinology and Metabolism 40,545–557.

Ying, S.-Y. (1988) Inhibins, activins and folbstatins: gonadal proteinsmodulating the secretion of FSH.Endocrine Reviews 9, 267–293.

Yuan, Q.X., Swerdloff, R.S. & Bhasin, S.(1988) Differential regulation of rat LH α and β subunits during the stimulatoryand downregulatory phases of GnRHaction. Endocrinology 122, 504–510.

Zhou, Z.-X., Wong, C.-I., Sar, M. & Wilson,E.M. (1994) The androgen receptor: anoverview. Recent Progress in HormoneResearch 49, 249–274.

Zmeili, S.M., Papavasiliou, S.S., Thorner,M.O. et al. (1986) Alpha and LH βsubunit mRNAs during the rat estrouscycle. Endocrinology 119, 1867–1869.

Zmuda, J.M., Cauley, J.A., Kriska, A. et al. (1997) Longitudinal relationbetween endogenous testosterone andcardiovascular disease risk factors in middle-aged men. A 13-year follow-up of former Multiple Risk FactorIntervention Trial participants. American Journal of Epidemiology 146,609–617.

Winter, J.S.D., Hughes, I.A., Reyes, F.L. &Faiman, C. (1976) Pituitary–gonadalrelations in infancy: patterns of serumgonadal steroid concentrations in manfrom birth to 2 years of age. Journal ofClinical Endocrinology and Metabolism 42,679–686.

Winters, S.J., Sherins, R.J. & Loriaux, D.L.(1979) Studies on the role of sex steroidsin the feedback control of gonadotropinconcentration in men. Journal of ClinicalEndocrinology and Metabolism 48,553–558.

Yamaji, T., Dierschke, D.J., Bhattacharya,A.N. & Knobil, E. (1992) The negativefeedback control by estradiol andprogesterone of LH secretion in theovariectormized rhesus monkey.Endocrinology 90, 771–778.

Yamamoto, M., Diebell, N.D. &Bogdanove, E.M. (1970) Analysis of initial and delayed effects oforchidectomy and ovariectomy onpituitary and serum LH levels in adultand immature rats. Endocrinology 86,1102–1111.

Page 322: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

306

Introduction

Physical exercise can be assimilated to a very com-plex physiological stimulus that challenges diverseaspects of the cellular function. Skeletal muscle isone of the tissues that respond to exercise by under-going a series of adjustments at the level of sev-eral of its components. The shortening velocity of skeletal muscles, the amount of force they are able togenerate and their capacity to resist fatigue areimportant properties closely related to athletic per-formances. Thanks to the high degree of malleabil-ity of different muscular parameters such as fibersize, fiber type composition and capillarization,skeletal muscles adapt adequately to changes im-posed by training. However, skeletal muscles willadapt differently to endurance and strength exer-cises, suggesting the existence of different sensingsystems. Therefore the adaptive process of skeletalmuscles to training can be viewed as orchestratedlocal and peripheral events where hormonal, mech-anical, metabolic and neural factors are key regu-latory signals. Changes in the rate of synthesis ofhormones and growth factors, and the expression of their receptors, are important signals involved inthe adaptive process allowing skeletal muscles tomeet the physiological demand of different types of physical activities. A brief summary of the roleplayed by some hormones and growth factors inmuscle hypertrophy, in the regulation of musclefiber phenotype and in the remodeling of capillarynetwork form the substance of this chapter.

The enlargement of skeletal muscles: the role myonuclei and satellite cells

Enhanced contractile protein synthesis is an un-equivocal condition for the increase in the size ofmuscle fibers in response to training. Both proteinsynthesis and degradation rates are altered duringthe enlargement of skeletal muscles (Goldberg et al.1975). The increase in muscle protein synthesisabove resting levels occurs very rapidly, between 1 and 4 h after the completion of a single bout ofexercise in humans (Wong & Booth 1990; Chesley et al. 1992; Biolo et al. 1995; Phillips et al. 1997). At the onset of muscle hypertrophy, increased proteinsynthesis correlates with an increase in RNA activ-ity (Laurent et al. 1978; Wong & Booth 1990). Thetranslation of mRNA is enhanced by factors whoseactivity is known to be regulated by their phos-phorylation state (Frederickson & Sonenberg 1993;Wada et al. 1996). Paralleling these changes, aminoacid transport into exercising muscles is also in-creased following training. This would theoreticallyenhance the availability of amino acids for newmuscle protein synthesis (Biolo et al. 1997).

Following this initial step of muscle fiber hyper-trophy, several lines of evidence indicate thatincreased RNA levels (rather than RNA activity)appear to be essential for muscle fibers to continueto hypertrophy. In this regard, increased amount ofmRNA can be attributed to either increased genetranscription per myonucleus or to increased num-ber of myonuclei. Adult muscle fibers contain hundreds of myonuclei, where each myonucleussustains the protein synthesis over a finite volume

Chapter 22

Hormonal and Growth Factor-RelatedMechanisms Involved in the Adaptation ofSkeletal Muscle to Exercise

FAWZI KADI

Page 323: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 307

source for the addition of new myonuclei intohypertrophied fibers (Moss & Leblond 1971;Schiaffino et al. 1976). Satellite cells are locatedbetween the basal lamina and the plasma mem-brane of muscle fibers (Mauro 1961); they are char-acterized by a high nuclear to cytoplasmic ratio, a well-developed Golgi apparatus, a prominentrough endoplasmic reticulum and a heterochrom-atic nucleus (Campion 1984). Several stimuli canactivate satellite cells, which then undergo prolifera-tion. The daughter cells then fuse with the under-lying adult muscle fiber providing new myonuclei.The involvement of satellite cell-derived myonucleiin fiber hypertrophy is further supported by experi-ments demonstrating that satellite cell activationand proliferation is required to support the enlarge-ment of muscles in animal models (Rosenblatt &Parry 1992; Rosenblatt et al. 1994).

In parallel with muscle fiber hypertrophy, it hasbeen shown that heavy resistance strength traininginduced a significant increase in the number ofsatellite cells in human skeletal muscles (Kadi 2000;Roth et al. 2001). A 46% increase in satellite cell fre-quency was reported in skeletal muscle of youngwomen after 10 weeks of strength training (Kadi &Thornell 2000). More recently, an increase in thenumber of satellite cells was found in skeletal mus-cles of a group of men aged between 70 and 80 yearsin response to endurance training (Charifi et al.2003a). Thus, satellite cells contribute to the acquisi-tion of new myonuclei and to the renewal of theirown pool (Bischoff 1994; Schultz & McCormick1994). Finally, the proliferation of satellite cells followed by the fusion of daughter cells togetherwould give rise to new muscle fibers (Kennedy et al.1988; Yamada et al. 1989; McCormick & Schultz 1992;Antonio & Gonyea 1993; Kadi & Thornell 1999). The newly formed muscle fibers would replacedamaged fibers or contribute to fiber hyperplasia ifthe number of newly generated fibers exceeds thenumber of injured fibers upon training (Fig. 22.1).

The effects of androgenic-anabolic steroids

The use of androgenic-anabolic steroids is accom-panied by a remarkable increase in muscle size andstrength in animal studies (Egginton 1987; Salmons

of cytoplasm, a concept called ‘nuclear domain’(Cheek 1985; Hall & Ralston 1989; Allen et al. 1999).It is important to note that although myonuclei arepost-mitotic, they are nonetheless able to sustain theenlargement of fibers up to a certain limit afterwhich the recruitment of new myonuclei becomesnecessary. In agreement with this statement, resultsfrom animal and human studies showed that thehypertrophy of skeletal muscle fibers was accom-panied by significant increases in the myonuclearnumber (Goldberg et al. 1975; Cabric & James 1983;Winchester & Gonyea 1992; Allen et al. 1995; Kadi2000). In well-trained humans, the number of myo-nuclei in hypertrophied skeletal muscle fibers ofpower lifters is higher than that of sedentary sub-jects, and a linear relationship between the numberof myonuclei and the cross-sectional area of musclefibers is found (Kadi et al. 1999a; Kadi 2000). Theaddition of new myonuclei to the enlarged musclefibers would play a role in the maintenance of a constant myonuclei/cytoplasm ratio, i.e. nucleardomain. The incorporation of additional myonucleiinto hypertrophying human muscle fibers wasreported both in young and elderly subjects (Hikidaet al. 1998; Kadi & Thornell 2000).

While the number of myonuclei per fiber isincreased during muscle fiber hypertrophy, the con-verse has also been documented in animal studies.A decrease in the number of myonuclei occurs as a result of an ongoing atrophy of muscle fibersinduced by spinal cord transection (Allen et al.1995), space flight (Allen et al. 1996) or hindlimb sus-pension (Hikida et al. 1997). Thus, the modulation of the myonuclear population seems to be of greatimportance in the regulation of fiber size. However,it is important to keep in mind that the enhancementof the myonuclear number would occur only whenthe transcriptional capacity of existing myonucleibecomes unable to support the enlargement of thefiber. Indeed, significant increases in myonuclearnumber have been observed in muscle fibers thathad hypertrophied by more than 26% (Cabric &James 1983; Allen et al. 1995; Roy et al. 1999; Kadi &Thornell 2000), but not by 6.8–15.5% (Giddings & Gonyea 1992).

As existing myonuclei in an adult muscle fiber are postmitotic, muscle satellite cells are the major

Page 324: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

308 chapter 22

1992). The administration of supra-physiologicaldoses of testosterone for 10 weeks in untrained and trained men produced a significant increase inmuscle strength and in the cross-sectional area of the quadriceps (Bhasin et al. 1996). Androgenic-anabolic steroids are known to increase the rate ofprotein synthesis and to promote muscle growthboth in vivo and in vitro (Powers & Florini 1975;Rogozkin 1979). In humans, long-term anabolicsteroids usage accentuates the degree of fiber hyper-trophy of muscle fibers in well-trained power-lifters(Kadi et al. 1999b). Skeletal muscles of power-lifterstaken anabolic steroids were characterized by both extremely large fibers and high myonuclearnumbers (Kadi et al. 1999b). Similarly, using animalmodels, it was also found that androgenic-anabolicsteroids mediate their myotrophic effect by enhan-cing the myonuclear content of muscles fibers and by increasing the number of muscle fibers (Galavazi& Szirmai 1971; Sassoon & Kelley 1986; Joubert &Tobin 1989; Joubert & Tobin 1995). Thus, androgenic-anabolic steroids would increase the myonuclearnumber to sustain the protein synthesis of extremely

hypertrophied muscle fibers (Kadi et al. 1999b). Amain mechanism by which androgenic-anabolicsteroids induce muscle hypertrophy is by activatingand inducing the proliferation of satellite cells,which subsequently incorporate into muscle fibersor fuse together to form new muscle fibers. In agree-ment with this statement is the immunohistochem-ical localization of androgen receptors in culturedsatellite cells indicating that anabolic steroids canact directly on muscle satellite cells (Doumit et al.1996).

Androgen receptors

Blockade of androgen receptors by oxendolone, an androgen receptor antagonist, suppressed thehypertrophy caused by exercise (Inoue et al. 1994).Although several factors are involved in the adapta-tion of muscle fibers to exercise, this experimentclearly shows that androgen receptors are import-ant mediators of the exercise-induced muscle fiberhypertrophy.

Androgen receptors belong to the family of ligand-responsive transcription regulators. When hormones bind to the receptor, it becomes activatedand the hormone-receptor complex is translocatedto the hormone responsive element within thenucleus. The binding to selective genes increases therates of transcription (Luke & Coffey 1994). Earlyreports indicated that androgen receptors werelocated in the cytosol of muscle fibers (Krieg 1976;Max et al. 1981). Using immunohistochemistry withspecific polyclonal and monoclonal antibodies, thenuclear location of androgen receptors has beenclearly demonstrated in nearly all tissues (Sar et al.1990; Takeda et al. 1990; Ruizeveld-De-Winter et al.1991; Kimura et al. 1993; Janssen et al. 1994). In skel-etal muscles, androgen receptors are expressed inmyonuclei (Takeda et al. 1990; Kimura et al. 1993;Dorlochter et al. 1994; Kadi et al. 2000b) and in satel-lite cells (Doumit et al. 1996). In normal restinghuman muscle fibers, androgen receptors are ex-pressed in some but not all myonuclei (Kadi et al.2000b), and differences in androgen receptor con-tent have been reported between human trapeziusand vastus lateralis muscles (Kadi 2000). Similarintermuscular differences in androgen receptor

Training, hormones and growth factors

Activated satellite cell

Dotter cells

1. Provide additional myonuclei2. Generate new muscle fibers or repair injured muscle fibers3. Enhance satellite cell pool

S

S

S SS S S

SS

S

SS

MN

MNMNMuscle fiber

Fig. 22.1 The effects of training, hormones and growthfactors on satellite cells. MN, myonucleus; S, satellite cell.

Page 325: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 309

content have also been demonstrated in frog skel-etal muscles (Dorlochter et al. 1994). In agreementwith the intermuscular differences in androgenreceptor content, it has been shown that the sensitiv-ity of guinea pig skeletal muscles to testosteronestimulation was greatest in the head and neckregion and gradually decreased from head to hind-quarters (Kochakian & Tillotson 1957). Intermuscu-lar differences in androgen receptor content mightwell reflect differences in embryological origins,nerve supply and functional requirements of differ-ent muscles.

Training influences the number of androgenbinding sites in skeletal muscles. The increase inandrogen receptors would lead to an enhancementof the sensitivity of muscles to circulating andro-gens. A significant increase in the number of andro-gen receptors has been shown to occur followingendurance and strength training, and electricalstimulation in animal studies (Inoue et al. 1993;Deschenes et al. 1994). The amplitude of changes in androgen receptor content following training is muscle dependent (Hickson & Kurowski 1986;Deschenes et al. 1994; Kadi et al. 2000b). Long-termstrength training is associated with changes inandrogen receptor-containing myonuclei in humantrapezius but not vastus lateralis muscle (Kadi et al.2000b). Similar differences in the regulation ofandrogen receptors following exercise exist betweenrat soleus, extensor digitorum longus, gastrocne-mius and plantaris muscles (Hickson & Kurowski1986; Salmons 1992; Inoue et al. 1993; Bricout et al.1994; Deschenes et al. 1994).

Deschenes et al. (1994) examined the effects ofendurance and resistance training on androgenreceptor content and receptor affinity to dihydro-testosterone in rat fast and slow skeletal muscles.Neither endurance nor resistance training inducedalterations in androgen receptor affinity to dihy-drotestosterone. Endurance training induced anenhancement in androgen binding capacity in theslow muscle whereas resistance training induced an enhancement in androgen binding capacity inthe fast muscle. Thus, alterations in androgen receptor content are not only muscle dependent,they also depend upon the type of physical activity(endurance/strength exercises). The changes in

androgen receptors following training occur rapidly.By 3 days of electrical stimulation, a 25% increase in the number of androgen receptors was reportedin the stimulated rat gastrocnemius muscle, andwas followed by a progressive hypertrophy of themuscle (Inoue et al. 1993).

The effects of androgenic-anabolic steroids onandrogen receptor content have also been invest-igated. In animal models and in cultured musclesatellite cells, it has been shown that androgenic-anabolic steroids may either up-regulate (Doumit et al. 1996) or down-regulate (Lin et al. 1993; Bricoutet al. 1994) androgen receptor content. Doumit et al.(1996) showed that the administration of testo-sterone enhanced androgen receptor immunore-activity in porcine satellite cell nuclei. In contrast,using radio-competition assay, it has been shownthat the concentration of androgen receptors wasdecreased following androgenic-anabolic steroidstreatment in rat soleus and extensor digitorumlongus muscles (Bricout et al. 1994). In fact, theeffects of androgenic-anabolic steroids on androgenreceptors might also be muscle dependent. Frogskeletal muscle fibers in the shoulder region havebeen shown to be more sensitive to testosteronethan fibers from other regions (Regnier & Herrera1993a, 1993b). It has also been shown that two rabbitskeletal muscles characterized by similar fiber size,fiber type composition, nerve and blood vessel sup-ply can differ greatly in their response to andro-genic-anabolic steroid treatment (Salmons 1992).Finally, long-term self-administration of androgenic-anabolic steroids in humans was associated withchanges in androgen receptor-containing myonu-clei in the trapezius but not in the vastus lateralis(Kadi et al. 2000b). Clearly, further studies are warranted to better understand the modulation ofandrogen receptors in skeletal muscles in responseto physiological and supra-physiological conditions.

Fibroblast growth factors

The fibroblast growth factor (FGF) family comprisesof ten members involved in different biologicalfunctions (Yamaguchi & Rossant 1995). Some FGFisoforms have been shown to play an important rolein the enlargement of muscle fibers in response to a

Page 326: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

310 chapter 22

physiological stimulus and others may exert theirmyotrophic role during the reparation of musclefibers following fiber damage. In this respect, FGF6is an important factor contributing to normal skel-etal muscle regeneration following injury (Floss et al. 1997). FGF6 would be involved in importantregenerative events such as the activation and pro-liferation of satellite cells and the expression ofimportant myogenic regulatory factors. FGF2 andFGF4, which are localized in the myofiber peripheralmatrix in adult skeletal muscles, are up-regulatedduring stretch mediated hypertrophy in an avianwing-weighting model (Mitchell et al. 1999). The up-regulation and specific cellular location of FGF2and FGF4 support their role in the activation andproliferation of satellite cells (Mitchell et al. 1999).Located in the same matrix as satellite cells, it ishypothesized that the release of these FGF isoformsfrom heparin components might be involved in thegeneration of new muscle fibers following training(Yamada et al. 1989). Alterations in the interactionFGF/heparan-sulfate proteoglycans in exercisingmuscles would modulate the availability of FGF tosatellite cells.

Investigating the mechanisms governing the con-version of a mechanical load into a skeletal musclegrowth response, Clarke & Feeback (1996) foundthat the release of FGF2 increases in parallel withincreased mechanical load in a tissue culture modelof differentiated human skeletal muscle cells. Thegrowth response was inhibited when the biologicactivity of FGF2 was neutralized (Clarke & Feeback1996). This experiment strongly suggests that FGF2release is an important autocrine mechanism fortransducing the stimulus of mechanical load into askeletal muscle growth response (Clarke & Feeback1996).

Insulin-like growth factors and receptor

Insulin-like growth factor (IGF) isoforms are pro-duced by many tissues and are important in bothembryonic and postnatal development. IGF-II is es-sential for normal embryonic development whereasIGF-I is important for both pre- and postnatalgrowth (DeChiara et al. 1990; Baker et al. 1993). Exer-cising skeletal muscles produce and utilize IGF-I

(Brahm et al. 1997), and IGF-I is considered as an important factor mediating the enlargement ofskeletal muscles in response to training. IGF-I is ableto stimulate satellite cells proliferation, differenti-ation and fusion (Dodson et al. 1985; Florini et al. 1991; Quinn et al. 1994; Goldspink, D.F. et al. 1995).Immunohistochemical studies showed that IGF-Iprotein is expressed in satellite cells and in myo-tubes of regenerating rat skeletal muscles ( Jennischeet al. 1987; Jennische 1989; Jennische & Matejka1992).

An increase in IGF-I immunoreactivity mostlywithin the muscle fibers was observed following an acute bout of eccentric exercise in rat tibialis anterior muscle (Yan et al. 1993). In humans, 7 daysof strenuous exercise induced an elevation of IGF-Iimmunoreactivity in the vastus lateralis muscle(Hellsten et al. 1996). The immunostaining for IGF-Iwas located on nuclei that might either representmyonuclei or satellite cells (Hellsten et al. 1996).Enhanced IGF-I mRNA and protein in parallel withthe hypertrophy of muscle fibers indicates that IGF-I is a key factor involved in the process of musclefiber enlargement via the recruitment of musclesatellite cells (Adams & Haddad 1996). Bamman et al. (2001) showed that IGF-I mRNA levels in-creased 48 h after the completion of a single bout of concentric and eccentric muscle loading in humanvastus lateralis muscle (the increase following ec-centric loading being more important). In the samestudy, a substantial increase in androgen receptormRNA was also found to occur following both con-centric and eccentric loading.

Advances in molecular biology allowed the discovery of new IGF-I isoforms (Yang et al. 1996;Goldspink, G. 1999; Hameed et al. 2002). Two maintypes of IGF-I expressed in skeletal muscles havebeen described. The first muscle isoform with a systemic mode of action is known as muscle liver-type L.IGF-I and is similar to the main liver IGF-I(Yang et al. 1996). The second muscle isoform, calledmechano growth factor (MGF) has been discoveredin skeletal muscles subjected to stretch and overloadand has an autocrine/paracrine action (Yang et al.1996). In response to stretch by immobilizing thehindlimb in the extended position, a substantialincrease in MGF was observed in rabbit extensor

Page 327: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 311

digitorum longus (Yang et al. 1996). Later, it hasbeen shown that both muscle L.IGF-I and MGFmRNAs were significantly increased in rabbitextensor digitorum longus following stretch andstretch combined with electrical stimulation at 10 Hzbut not after electrical stimulation alone (McKoy et al. 1999). These experiments clearly showed thatIGF-I isoforms mediate the enlargement of skeletalmuscles mainly in response to increased mechanicalload. The involvement of MGF in fiber hypertrophyhas been investigated in young and elderly subjects.A significant increase in MGF mRNA occured in theyoung but not in the elderly subjects following 10sets of six repetitions of single-legged knee extensorexercise at 80% of 1-repetition maximum (1-RM)(Hameed et al. 2003). It was concluded that thereduced MGF response to high resistance exercise in elderly subjects might indicate an age-relateddesensitivity to mechanical loading.

The administration of IGF-I has been consideredfor the treatment of various neuromuscular diseasescharacterized by muscular atrophy. In this respect,IGF-I has been successfully used to prevent thedevelopment of steroid-induced muscle atrophy(Kanda et al. 1999). At the cellular level, it has beenshown that daily growth hormone/IGF-I adminis-tration combined with functional overload of ratsoleus muscle results in the enlargement of musclesand a concomitant enhancement of the myonuclearnumber of muscle fibers (McCall et al. 1998).

IGF-I and IGF-II promote their growth effectsthrough IGF-I receptor. Mice with disturbed IGF-Ireceptor die soon after birth (Baker et al. 1993). Thelack of IGF-I receptor induces severe hypoplasia inmice suggesting that IGF-I receptor is essential forthe establishment of a mature skeletal muscle (Bakeret al. 1993). While functional inactivation of the IGF-I receptor induced a marked muscle hypoplasiaand a decrease in MyoD and myogenin levels (twoimportant members of the myogenic regulatory factors) (Fernandez et al. 2002), the opposite occurswhen the IGF-I receptor is overexpressed (Quinn et al. 1994). Therefore, IGF-I receptor is currentlyconsidered as a key regulator of muscle mass via itsaction on muscle-specific genes. A single acute boutof exercise has been shown to be accompanied by asignificant increase in the IGF-I receptor binding

capacity and affinity as well as in IGF-I receptormRNA in rat skeletal muscle (Willis et al. 1997).Similarly, long-term training induced a significantincrease in IGF-I and insulin receptor content (Williset al. 1998). Altogether, these findings strongly sug-gest that IGF-I and IGF-I receptors are essentialmediators of muscle fiber hypertrophy.

Capillarization of skeletal muscles

Blood vessels in the skeletal muscles form a rich net-work of capillaries around muscle fibers. Capillariesconsist of a single layer of endothelial cells sur-rounded by a luminal glycocalyx and an ablum-inal basement membrane. Located at the end of the cardiorespiratory system, the capillary networkplays an important role in nutrients, and oxygenand carbon dioxide exchange with muscle fibers. Inresponse to endurance training, a remodeling of thecapillary bed has been shown to occur in differenthuman skeletal muscles (Andersen, P. & Henriksson1977; Hudlicka et al. 1992; Wang et al. 1993; Kadi et al.2000a; Charifi et al. 2003b).

Vascular endothelial growth factor

Vascular endothelial growth factor (VEGF) is a heparin-binding endothelial cell-specific mitogenmediating angiogenesis in different tissues. Bothexercise and hypoxia can cause an increase in VEGFmRNA in human skeletal muscle (Gustafsson et al.1999; Richardson et al. 1999). VEGF mRNA increasedin response to a single bout of exercise at a workload corresponding to 50% of peak work load innormal healthy subjects and in patients with chronicrenal failure (Wagner et al. 2001). The receptors forVEGF (flt-1 and flk-1) are also up-regulated follow-ing both exercise and hypoxia (Takagi et al. 1996;Gerber et al. 1997; Olfert et al. 2001). Thus, VEGF andits receptors are involved in the densification of thecapillary network in response to physical activity.There is a strong increase in VEGF mRNA followinga single bout of exercise in untrained human skel-etal muscles. VEGF response is attenuated in skeletalmuscle of trained subjects (Richardson et al. 2000).This might reflect an initial intense proliferation ofcapillaries in untrained muscles occurring at the

Page 328: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

312 chapter 22

beginning of a training program, followed by aperiod where the enhancement of the capillariza-tion occurs at a slower rate and requires high dosesof training as the training status of muscles isimproved.

Among factors controlling the expression ofVEGF, hypoxia inducible factor 1 (HIF-1) subunit iscurrently considered as a major regulatory factor.The effects of short-term exercise training on VEGFand HIF-1 have been studied in eight healthy males.Although VEGF levels increased following seventraining sessions, no changes in the levels of HIF-1mRNA subunits were found (Gustafsson et al. 2002).This might indicate that an increase in HIF-1 mRNAis not the only factor involved in the regulation of training-induced VEGF enhancement. However,the intervention of different factors controlling theremodeling of the capillary network probably oc-curs during serial transient time frames making itdifficult to assess their importance unless the wholeadaptive process is monitored.

FGFs are also believed to play a role in skeletalmuscle angiogenesis. However, recent studies sug-gest that their contribution to angiogenesis is lessimportant than that played by VEGF (Richardson et al. 2000; Wagner et al. 2001). At present, althoughit is suggested that VEGF is the most importantangiogenic factor involved in the adaptation of the capillary network in human skeletal muscles,further studies are needed to advance knowledgeabout the importance of all angiogenic factors inregulating skeletal muscle angiogenesis.

Contractile properties of muscle fibers

The existence of numerous types of fibers makesskeletal muscle a very heterogeneous tissue able toperform various functional capabilities. Immuno-histochemical and biochemical analysis of skeletalmuscles revealed that this diversity in muscle fibertypes reflects a broad spectrum of myosin isoforms.The myosin is the molecule primarily responsible,along with actin, for muscle contraction. The myosinmolecule is made up of two heavy chains (MyHC)and four light chains (MyLC) (Schiaffino & Reggiani1996; Pette & Staron 1997). The heavy chain portion

of the myosin molecule exists in multiple isoformsand represents a major determinant of the force-velocity properties of muscle fibers. The four mostimportant MyHC isoforms expressed in adult skel-etal muscle fibers are: MyHC Iβ, MyHC IIA, MyHCIIX/IID and MyHC IIB. Each isoform is charac-terized by a specific shortening velocity and forceproduction. Fibers containing MyHC I have a slowcontracting velocity and produce less force thanfibers containing MyHC IIA, IIX and IIB. Withinmuscle fibers containing fast MyHCs, the fastestand strongest are those containing MyHC IIB fol-lowed by fibers expressing MyHC IIX and MyHCIIA (Bottinelli et al. 1994a, 1994b).

The contractile properties of skeletal muscles areable to undergo significant changes in response toexercise. It is generally accepted that endurancetraining results in a fast to slow transition of MyHCisoforms (Baumann et al. 1987; Schaub et al. 1989),whereas strength training causes an increase inMyHC IIA and a decrease in MyHC IIX (Staron et al.1991; Adams et al. 1993; Andersen, J.L. et al. 1994; Fry et al. 1994; Kraemer et al. 1995; Kadi & Thornell 1999;Andersen, J.L. & Aagaard 2000). It is also suggestedthat muscle fibers containing MyHC IIX are seldomrecruited in the normal daily activities of most people.If they become more recruited, as it is the case dur-ing training, they are converted into fibers contain-ing MyHC IIA (type IIA fibers being more fatigueresistant than type IIX fibers) (Goldspink, G. et al.1991; Staron et al. 1991; Kraemer et al. 1995). Duringendurance or strength training, the hormonal envir-onment of skeletal muscles is greatly disturbed andthese alterations are powerful signals able to triggerchanges in myosin expression in exercising muscles.

Effects of testosterone

In some animal studies, a fast-to-slow shift of MyHCisoforms has been observed following androgenic-anabolic steroid treatment (Fritzsche et al. 1994;Czesla et al. 1997). An increase in fibers containingMyHC IIA and a decrease in fibers containingMyHC IIB has been reported in several skeletalmuscles in rodents in response to androgenic-anabolic steroid administration (Egginton 1987;

Page 329: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 313

Dimauro et al. 1992). In contrast, an androgenicsteroid-induced decrease in MyHC IIA-containingfibers in favor of MyHC IIB-containing fibers hasalso been reported (Kelly et al. 1985; Lyons et al.1986; Salmons 1992). These results suggest that theeffects of androgenic-anabolic steroids on the con-tractile properties might be muscle-dependant andmight also vary between species. In fact, there isother evidence indicating that androgenic-anabolicsteroids have no significant effects on muscle fibertype composition. For example, muscle overload inanimal experiments induces an increase in theexpression of slow MyHC I, and the addition ofandrogenic-anabolic steroids did not alter the patternof MyHC expression (Boissonneault et al. 1987). Like-wise, the administration of androgenic-anabolicsteroids did not modify the slow-to-fast shift inMyHC isoforms caused by hindlimb suspension ex-periments (Tsika et al. 1987). Finally, in well-trainedpower-lifters, there were no differences in trapeziusMyHC composition between androgenic-anabolicsteroid users and non-users (Kadi et al. 1999b).

Effects of estrogen

It is well-known that a reduction in force productionoccurs at menopause (Greeves et al. 1999; Dionne et al. 2000; Meeuwsen et al. 2000). At the cellularlevel, it has been shown that ovariectomy is asso-ciated with a fast-to-slow shift in MyHC isoformsand a reduction in the level of spontaneous runningactivity in rats (Kadi et al. 2002). MyHC changestend to follow a general pathway of sequential transition in the order MyHC I ← IIA ← IIX ← IIB. This result can be interpreted as an overall transi-tion in the expression of fast isoforms towardsslower isoforms, a specific up-regulation of theslower isoforms of MyHC genes, or a specific down-regulation of genes coding for fast MyHC isoforms following ovariectomy. When ovariec-tomized animals are allowed to run and are treatedwith estrogen, there were no alterations in MyHCcomposition (Kadi et al. 2002). Therefore, it can be suggested that physical activity together withestrogen treatment may help to maintain musclecharacteristics of both slow and fast muscles.

Effects of growth hormone

It has been reported that growth hormone admin-istration induced an increase in MyHC IIX in thevastus lateralis of healthy elderly men (Lange et al. 2002). The shift in MyHC isoforms towardsMyHC IIX has been interpreted as a change into amore youthful MyHC composition as a decrease in MyHC IIX usually accompanies aging in thismuscle group (Lange et al. 2002). In contrast, it has been shown that the amount of MyHC IIX ingrowth hormone-deficient patients was higher thanin a normal control population (Daugaard et al.1999). Furthermore, treatment of growth hormone-deficient patients with recombinant human growthhormone for 6 months had no effect on MyHC com-position (Daugaard et al. 1999). Similarly, it has beenshown that growth hormone treatment significantlyincreased the cross-sectional area of rat type II fibersin the soleus muscle with no significant effect onfiber type composition (Aroniadou-Anderjaska et al.1996). Whether increased concentration of growthhormone is associated with a slow-to-fast shift ofmyosin isoforms remains to be further investigated.

Effects of thyroid hormones

Thyroid hormones exert a powerful action in theregulation of MyHC composition of skeletal mus-cles (D’Albis & Butler-Browne 1993). Larsson & Yu (1997) showed that the regulation of MyHC isoforms in rat skeletal muscle by thyroid hormoneis gender and muscle specific. Administration of3.5.3’-triiodothyronine (T3) causes a down-regula-tion of MyHC I and up-regulation of MyHC IIA inmale and female soleus, whereas the up-regulationof MyHC IIX is observed only in male muscles(Larsson & Yu 1997). Treatment with T3 induces no changes in MyHC isoforms in male extensor dig-itorum longus (EDL). The same treatment induces asignificant transition from MyHC IIA to MyHC IIB infemale EDL (Larsson & Yu 1997). Altogether, theseresults show that the contractile properties of skeletalmuscles are under the control of several hormonesand growth factors, and that changes in the hormonalenvironment of skeletal muscles during exercise are

Page 330: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

314 chapter 22

partly responsible for the adjustment of muscle phenotype to the physiological demand. It is nowbecoming clear that changes in muscle structure andfunction in response to changes in the hormonalenvironment can be muscle and gender dependent.

Conclusion

This paper outlined only a few aspects related torole of specific hormones and growth factors in theregulation of some important muscular parametersresponsible for the athletic performance. This area

of muscle physiology has just begun to develop and there is still much to be discovered in order tobetter understand the different signals involved inthe various adaptive events occurring in skeletalmuscles in response to different forms of physicalactivities. The delineation of the different steps ofthe adaptation of muscles to exercise would providethe basis for the conception of individualized exer-cise prescriptions to optimize the quality of train-ing programs both in well-trained and sedentaryhealthy populations as well as in populations suf-fering from specific diseases.

References

Adams, G.R. & Haddad, F. (1996) Therelationships among IGF-1, DNAcontent, and protein accumulationduring skeletal muscle hypertrophy.Journal of Applied Physiology 81,2509–2516.

Adams, G.R., Hather, B.M., Baldwin, K.M.& Dudley, G.A. (1993) Skeletal musclemyosin heavy chain composition andresistance training. Journal of AppliedPhysiology 74, 911–915.

Allen, D.L., Monke, S.R., Talmadge, R.J.,Roy, R.R. & Edgerton, V.R. (1995)Plasticity of myonuclear number in hypertrophied and atrophiedmammalian skeletal muscle fibers.Journal of Applied Physiology 78,1969–1976.

Allen, D.L., Yasui, W., Tanaka, T. et al.(1996) Myonuclear number and myosinheavy chain expression in rat soleussingle muscle fibers after spaceflight.Journal of Applied Physiology 81, 145–151.

Allen, D.L., Roy, R.R. & Edgerton, V.R.(1999) Myonuclear domains in muscleadaptation and disease. Muscle and Nerve22, 1350–1360.

Andersen, J.L. & Aagaard, P. (2000)Myosin heavy chain IIX overshoot inhuman skeletal muscle. Muscle and Nerve23, 1095–1104.

Andersen, J.L., Klitgaard, H. & Saltin, B.(1994) Myosin heavy chain isoforms insingle fibres from m. vastus lateralis ofsprinters: influence of training. ActaPhysiologica Scandinavica 151, 135–142.

Andersen, P. & Henriksson, J. (1977)Capillary supply of the quadricepsfemoris muscle of man: adaptiveresponse to exercise. Journal of Physiology270, 677–690.

Antonio, J. & Gonyea, W.J. (1993) Skeletalmuscle fiber hyperplasia. Medicine andScience in Sports and Exercise 25,1333–1345.

Aroniadou-Anderjaska, V., Lemon, P.W.,Gilloteaux, J. (1996) Effects of exogenousgrowth hormone on skeletal muscle ofyoung female rats. Tissue and Cell 28,719–724.

Baker, J., Liu, J.P., Robertson, E.J. &Efstratiadis, A. (1993) Role of insulin-like growth factors in embryonic andpostnatal growth. Cell 75, 73–82.

Bamman, M.M., Shipp, J.R., Jiang, J. et al. (2001) Mechanical load increasesmuscle IGF-I and androgen receptormRNA concentrations in humans.American Journal of Physiology 280,383–390.

Baumann, H., Jaggi, M., Soland, F.,Howald, H. & Schaub, M.C. (1987)Exercise training induces transitions of myosin isoform subunits withinhistochemically typed human musclefibres. Pflugers Archiv 409, 349–360.

Bhasin, S., Storer, T.W., Berman, N. et al.(1996) The effects of supraphysiologicdoses of testosterone on muscle size andstrength in normal men. New EnglandJournal of Medicine 335, 1–7.

Biolo, G., Fleming, R.Y., Maggi, S.P. &Wolfe, R.R. (1995) Transmembranetransport and intracellular kinetics ofamino acids in human skeletal muscle.American Journal of Physiology 268,E75–E84.

Biolo, G., Tipton, K.D., Klein, S. & Wolfe,R.R. (1997) An abundant supply ofamino acids enhances the metaboliceffect of exercise on muscle protein.American Journal of Physiology 273,E122–E129.

Bischoff, R., 1994. The satellite cell andmuscle regeneration. In: Myology (Engel,A.G., Franzini-Armstrong, C., eds.).McGraw-Hill, New York: 97–117.

Boissonneault, G., Gagnon, J., Ho-Kim,M.A. & Tremblay, R.R. (1987) Lack ofeffect of anabolic steroids on specificmRNAs of skeletal muscle undergoingcompensatory hypertrophy. Molecularand Cellular Endocrinology 51, 19–24.

Bottinelli, R., Betto, R., Schiaffino, S. &Reggiani, C. (1994a) Maximumshortening velocity and coexistence ofmyosin heavy chain isoforms in singleskinned fast fibres of rat skeletal muscle.Journal of Muscle Research and CellMotility 15, 413–419.

Bottinelli, R., Betto, R., Schiaffino, S. & Reggiani, C. (1994b) Unloadedshortening velocity and myosin heavychain and alkali light chain isoformcomposition in rat skeletal muscle fibres.Journal of Physiology 478, 341–349.

Brahm, H., Piehl-Aulin, K., Saltin, B. &Ljunghall, S. (1997) Net fluxes overworking thigh of hormones, growthfactors and biomarkers of bonemetabolism during short lastingdynamic exercise. Calcified TissueInternational 60, 175–180.

Bricout, V., Germain, P., Serrurier, B. & Guezennec, C. (1994) Changes intestosterone muscle receptors: effects ofan androgen treatment on physicallytrained rats. Cellular and MolecularBiology 40, 291–294.

Cabric, M. & James, N.T. (1983)Morphometric analyses on the musclesof exercise trained and untrained dogs.American Journal of Anatomy 166,359–368.

Campion, D.R. (1984) The muscle satellite

Page 331: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 315

cell: a review. International Review ofCytology 87, 225–251.

Charifi, N., Kadi, F., Feasson, L. & Denis, C.(2003a) Effects of endurance training onsatellite cell frequency in skeletal muscleof old men. Muscle and Nerve 28, 87–92.

Charifi, N., Kadi, F., Feasson, L. et al.(2003b) Enhancement of microvesseltortuosity in the vastus lateralis muscleof old men in response to endurancetraining. Journal of Physiology 554, 559–569.

Cheek, D.B. (1985) The control of cell mass and replication. The DNA unit: a personal 20-year study. Early HumanDevelopment 12, 211–239.

Chesley, A., MacDougall J.D.,Tarnopolsky, M.A., Atkinson, S.A. &Smith, K. (1992) Changes in humanmuscle protein synthesis after resistanceexercise. Journal of Applied Physiology 73,1383–1388.

Clarke, M.S. & Feeback, D.L. (1996)Mechanical load induces sarcoplasmicwounding and FGF release indifferentiated human skeletal musclecultures. FASEB Journal 10, 502–509.

Czesla, M., Mehlhorn, G., Fritzsche, D. &Asmussen, G. (1997) Cardiomyoplasty:improvement of muscle fibre typetransformation by an anabolic steroid(metenolone). Journal of Molecular andCellular Cardiology 29, 2989–2996.

D’albis, A. & Butler-Browne, G. (1993) Thehormonal control of myosin isoformexpression in skeletal muscle ofmammals: a review. Basic and AppliedMyology 3(1), 7–16.

Daugaard, J.R., Bramnert, M., Manhem, P.et al. (1999) Effect of 6 months of GHtreatment on myosin heavy chaincomposition in GH-deficient patients.European Journal of Endocrinology 141,342–349.

DeChiara, T.M., Efstratiadis, A. &Robertson, E.J. (1990) A growth-deficiency phenotype in heterozygousmice carrying an insulin-like growthfactor II gene disrupted by targeting.Nature 345, 78–80.

Deschenes, M.R., Maresh, C.M.,Armstrong, L.E. et al. (1994) Enduranceand resistance exercise induce musclefiber type specific responses in androgenbinding capacity. Journal of SteroidBiochemistry and Molecular Biology 50,175–179.

Dimauro, J., Balnave, R.J. & Shorey, C.D.(1992) Effects of anabolic steroids andhigh intensity exercise on rat skeletalmuscle fibres and capillarization. Amorphometric study. European Journal of Applied Physiology 64, 204–212.

Galavazi, G. & Szirmai, J.A. (1971)Cytomorphometry of skeletal muscle:the influence of age and testosterone onthe rat m. levator ani. Zeitschrift furZellforschung und MikroskopischeAnatomie 121, 507–530.

Gerber, H.P., Condorelli, F., Park, J. & Ferrara, N. (1997) Differentialtranscriptional regulation of the twovascular endothelial growth factorreceptor genes. Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia. Journalof Biological Chemistry 272, 23 659–23 667.

Giddings, C.J. & Gonyea, W.J. (1992)Morphological observations supportingmuscle fiber hyperplasia followingweight-lifting exercise in cats.Anatomical Record 233, 178–195.

Goldberg, A.L., Etlinger, J.D., Goldspink,D.F. & Jablecki, C. (1975) Mechanism ofwork-induced hypertrophy of skeletalmuscle. Medicine and Science in Sports 7,185–198.

Goldspink, D.F., Cox, V.M., Smith, S.K.et al. (1995) Muscle growth in responseto mechanical stimuli. American Journalof Physiology 268, 288–297.

Goldspink, G. (1999) Changes in musclemass and phenotype and the expressionof autocrine and systemic growth factorsby muscle in response to stretch andoverload. Journal of Anatomy 194,323–334.

Goldspink, G., Scutt, A., Martindale, J. et al. (1991) Stretch and force generationinduce rapid hypertrophy and myosinisoform gene switching in adult skeletalmuscle. Biochemical Society Transactions19, 368–373.

Greeves, J.P., Cable, N.T., Reilly, T. & Kingsland, C. (1999) Changes inmuscle strength in women following themenopause: a longitudinal assessmentof the efficacy of hormone replacementtherapy. Clinical Science (London,England) 97(1), 79–84.

Gustafsson, T., Puntschart, A., Kaijser, L.,Jansson, E. & Sundberg, C.J. (1999)Exercise-induced expression ofangiogenesis-related transcription andgrowth factors in human skeletalmuscle. American Journal of Physiology276, 679–685.

Gustafsson, T., Knutsson, A., Puntschart,A. et al. (2002) Increased expression ofvascular endothelial growth factor inhuman skeletal muscle in response toshort-term one-legged exercise training.Pflugers Archiv 444, 752–759.

Hall, Z.W. & Ralston, E. (1989) Nucleardomains in muscle cells. Cell 59, 771–772.

Dionne, I.J., Kinaman, K.A. & Poehlman,E.T. (2000) Sarcopenia and musclefunction during menopause andhormone-replacement therapy. Journal ofNutrition, Health and Aging 4(3), 156–161.

Dodson, M.V., Allen, R.E. & Hossner, K.L.(1985) Ovine somatomedin,multiplication-stimulating activity, andinsulin promote skeletal muscle satellitecell proliferation in vitro. Endocrinology117, 2357–2363.

Dorlochter, M., Astrow, S.H. & Herrera,A.A. (1994) Effects of testosterone on a sexually dimorphic frog muscle:repeated in vivo observations andandrogen receptor distribution. Journalof Neurobiology 25, 897–916.

Doumit, M.E., Cook, D.R. & Merkel, R.A.(1996) Testosterone up-regulatesandrogen receptors and decreasesdifferentiation of porcine myogenicsatellite cells in vitro. Endocrinology 137,1385–1394.

Egginton, S. (1987) Effects of an anabolichormone on striated muscle growth and performance. Pflugers Archiv 410,349–355.

Fernandez, A.M., Dupont, J., Farrar, R.P. et al. (2002) Muscle-specific inactivationof the IGF-I receptor inducescompensatory hyperplasia in skeletalmuscle. Journal of Clinical Investigation109, 347–355.

Florini, J.R., Ewton, D.Z. & Roof, S.L.(1991) Insulin-like growth factor-Istimulates terminal myogenicdifferentiation by induction ofmyogenin gene expression. MolecularEndocrinology (Baltimore, Md.) 5, 718–724.

Floss, T., Arnold, H.H. & Braun, T. (1997) A role for FGF-6 in skeletal muscleregeneration. Genes and Development 11,2040–2051.

Frederickson, R. & Sonenberg, N., 1993.eIF-4E phosphorylation and theregulation of protein synthesis. In: Translational Regulation of GeneExpression (Ilan, J., ed.). Plenum Press,New York: 143–162.

Fritzsche, D., Krakor, R., Asmussen, G. et al. (1994) Effect of an anabolic steroid(Metenolon) on contractile performanceof the chronically stimulated latissimusdorsi in sheep. European Journal of Cardio-thoracic Surgery 8, 214–219.

Fry, A.C., Allemeier, C.A. & Staron, R.S.(1994) Correlation between percentagefiber type area and myosin heavy chaincontent in human skeletal muscle.European Journal of Applied Physiology 68,246–251.

Page 332: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

316 chapter 22

Hameed, M., Harridge, S.D. & Goldspink,G. (2002) Sarcopenia and hypertrophy: a role for insulin-like growth factor-1 inaged muscle? Exercise and Sport SciencesReviews 30, 15–19.

Hameed, M., Orrell, R.W., Cobbold, M.,Goldspink, G. & Harridge, S.D. (2003)Expression of IGF-I splice variants inyoung and old human skeletal muscleafter high resistance exercise. Journal ofPhysiology 547, 247–254.

Hellsten, Y., Hansson, H.A., Johnson, L.,Frandsen, U. & Sjodin, B. (1996)Increased expression of xanthineoxidase and insulin-like growth factor I(IGF-I) immunoreactivity in skeletalmuscle after strenuous exercise inhumans. Acta Physiologica Scandinavica157, 191–197.

Hickson, R.C. & Kurowski, T.G. (1986)Anabolic steroids and training. Clinics in Sports Medicine 5, 461–469.

Hikida, R.S., Van-Nostran, S., Murray, J.D.et al. (1997) Myonuclear loss in atrophiedsoleus muscle fibers. Anatomical Record247, 350–354.

Hikida, R.S., Walsh, S., Barylski, N. et al.(1998) Is hypertrophy limited in elderlymuscle fibers? A comparison of elderlyand young strength-trained men. Basicand Applied Myology 8(6), 419–427.

Hudlicka, O., Brown, M. & Egginton, S.(1992) Angiogenesis in skeletal andcardiac muscle. Physiological Reviews 72,369–417.

Inoue, K., Yamasaki, S., Fushiki, T. et al.(1993) Rapid increase in the number ofandrogen receptors following electricalstimulation of the rat muscle. EuropeanJournal of Applied Physiology 66, 134–140.

Inoue, K., Yamasaki, S., Fushiki, T., Okada,Y. & Sugimoto, E. (1994) Androgenreceptor antagonist suppresses exercise-induced hypertrophy of skeletal muscle.European Journal of Applied Physiology 69,88–91.

Janssen, P.J., Brinkmann, A.O., Boersma,W.J. & Van-Der-Kwast, T.H. (1994)Immunohistochemical detection of theandrogen receptor with monoclonalantibody F39.4 in routinely processed,paraffin-embedded human tissues aftermicrowave pre-treatment. Journal ofHistochemistry and Cytochemistry 42,1169–1175.

Jennische, E. (1989) Sequentialimmunohistochemical expression ofIGF-I and the transferrin receptor inregenerating rat muscle in vivo. ActaEndocrinologica 121, 733–738.

Jennische, E. & Matejka, G.L. (1992) IGF-Ibinding and IGF-I expression in

ovariectomy. Journal of Muscle Researchand Cell Motility 23, 335–339.

Kanda, F., Takatani, K., Okuda, S.,Matsushita, T. & Chihara, K. (1999)Preventive effects of insulin-like growthfactor-I on steroid-induced muscleatrophy. Muscle and Nerve 22, 213–217.

Kelly, A., Lyons, G., Gambki, B. &Rubinstein, N. (1985) Influences oftestosterone on contractile proteins of the guinea pig temporalis muscle.Advances in Experimental Medicine andBiology 182, 155–168.

Kennedy, J.M., Eisenberg, B.R., Reid, S.K.,Sweeney, L.J. & Zak, R. (1988) Nascentmuscle fiber appearance in overloadedchicken slow-tonic muscle. AmericanJournal of Anatomy 181, 203–215.

Kimura, N., Mizokami, A., Oonuma, T.,Sasano, H. & Nagura, H. (1993)Immunocytochemical localization ofandrogen receptor with polyclonalantibody in paraffin-embedded humantissues [see comments]. Journal ofHistochemistry and Cytochemistry. 41,671–678.

Kochakian, C.D. & Tillotson, C. (1957)Influence of several C19-steroids on thegrowth of individual muscles of theguinea pig. Endocrinology 60, 607–618.

Kraemer, W.J., Patton, J.F., Gordon, S.E. et al. (1995) Compatibility of high-intensity strength and endurancetraining on hormonal and skeletalmuscle adaptations. Journal of AppliedPhysiology 78, 976–989.

Krieg, M. (1976) Characterization of theandrogen receptor in the skeletal muscleof the rat. Steroids 28, 261–274.

Lange, K.H., Andersen, J.L., Beyer, N. et al. (2002) GH administration changesmyosin heavy chain isoforms in skeletalmuscle but does not augment musclestrength or hypertrophy, either alone or combined with resistance exercisetraining in healthy elderly men. Journalof Clinical Endocrinology and Metabolism87, 513–523.

Larsson, L. & Yu, F. (1997) Gender-relateddifferences in the regulatory influence of thyroid hormone on the expression ofmyosin isoforms in young and old rats.Acta Physiologica Scandinavica 159, 81–89.

Laurent, G., Sparrow, M. & Millward, D.(1978) Turnover of muscle protein in the fowl. Changes in rates of proteinsynthesis and beackdown duringhypertrophy of the anterior andposterior latissimus dorsi muscle.Biochemical Journal 176, 407–417.

Lin, M.C., Rajfer, J., Swerdloff, R.S. &Gonzalez-Cadavid, N.F. (1993)

regenerating muscle of normal andhypophysectomized rats. ActaPhysiologica Scandinavica 146, 79–86.

Jennische, E., Skottner, A. & Hansson, H.A.(1987) Satellite cells express the trophicfactor IGF-I in regenerating skeletalmuscle. Acta Physiologica Scandinavica129, 9–15.

Joubert, Y. & Tobin, C. (1989) Satellite cellproliferation and increase in the numberof myonuclei induced by testosterone in the levator ani muscle of the adultfemale rat. Developmental Biology 131,550–557.

Joubert, Y. & Tobin, C. (1995) Testosteronetreatment results in quiescent satellitecells being activated and recruited intocell cycle in rat levator ani muscle.Developmental Biology 169, 286–294.

Kadi, F. (2000) Adaptation of humanskeletal muscle to training and anabolicsteroids. Acta Physiologica Scandinavica646, 1–52.

Kadi, F. & Thornell, L.E. (1999) Trainingaffects myosin heavy chain phenotype in the trapezius muscle of women.Histochemistry and Cell Biology 112, 73–78.

Kadi, F. & Thornell, L.E. (2000)Concomitant increases in myonuclearand satellite cell content in femaletrapezius muscle following strengthtraining. Histochemistry and Cell Biology113, 99–103.

Kadi, F., Eriksson, A., Holmner, S., Butler-Browne, G.S. & Thornell, L.E. (1999a)Cellular adaptation of the trapeziusmuscle in strength-trained athletes.Histochemistry and Cell Biology 111,189–195.

Kadi, F., Eriksson, A., Holmner, S. &Thornell, L.-E. (1999b) Effects of anabolicsteroids on the muscle cells of strength-trained athletes. Medicine and Science inSports and Exercise 31, 1528–1534.

Kadi, F., Ahlgren, C., Waling, K., Sundelin,G. & Thornell, L.E. (2000a) The effects of different training programs on thetrapezius muscle of women with work-related neck and shoulder myalgia. ActaNeuropathologica 100, 253–258.

Kadi, F., Bonnerud, P., Eriksson, A. &Thornell, L.-E. (2000b) The expression ofandrogen receptors in human neck andlimb muscles: effects of training and self-administration of androgenic-anabolicsteroids. Histochemistry and Cell Biology113, 25–29.

Kadi, F., Karlsson, C., Larsson, B. et al.(2002) The effects of physical activityand estrogen treatment on rat fast andslow skeletal muscles following

Page 333: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones, skeletal muscle and exercise 317

Testosterone down-regulates the levelsof androgen receptor mRNA in smoothmuscle cells from the rat corporacavernosa via aromatization toestrogens. Journal of Steroid Biochemistryand Molecular Biology 45, 333–343.

Luke, M. & Coffey, D., 1994. The maleaccessory tissues: structure, androgen,and physiology. In: The Physiology ofReproduction (Knobil, E. & Neill, J., eds.). Raven Press, New York:1435–1488.

Lyons, G.E., Kelly, A.M. & Rubinstein,N.A. (1986) Testosterone-inducedchanges in contractile protein isoformsin the sexually dimorphic temporalismuscle of the guinea pig. Journal ofBiological Chemistry 261, 13 278–13 284.

McCall, G., Allen, D., Linderman, J. et al.(1998) Maintenance of myonucleardomain size in rat soleus after overloadand growth hormone/IGF-I treatment.Journal of Applied Physiology 84,1407–1412.

McCormick, K.M. & Schultz, E. (1992)Mechanisms of nascent fiber formationduring avian skeletal musclehypertrophy. Developmental Biology 150,319–334.

McKoy, G., Ashley, W., Mander, J. et al.(1999) Expression of insulin growthfactor-1 splice variants and structuralgenes in rabbit skeletal muscle inducedby stretch and stimulation. Journal ofPhysiology 516, 583–592.

Mauro, A. (1961) Satellite cell of skeletalmuscle fibers. Journal of Biophysical andBiochemical Cytology 9, 493–495.

Max, S.R., Mufti, S. & Carlson, B.M. (1981) Cytosolic androgen receptor inregenerating rat levator ani muscle.Biochemical Journal 200, 77–82.

Meeuwsen, I.B., Samson, M.M. & Verhaa, H.J. (2000) Evaluation of theapplicability of HRT as a preservative of muscle strength in women. Maturitas36(1), 49–61.

Mitchell, P., Steenstrup, T. & Hannon, K.(1999) Expression of fibroblast growthfactor family during postnatal skeletalmuscle hypertrophy. Journal of AppliedPhysiology 86, 313–319.

Moss, F.P. & Leblond, C.P. (1971) Satellitecells as the source of nuclei in muscles of growing rats. Anatomical Record 170,421–435.

Olfert, I.M., Breen, E.C., Mathieu-Costello,O. & Wagner, P.D. (2001) Chronichypoxia attenuates resting and exercise-induced VEGF, flt-1, and flk-1 mRNAlevels in skeletal muscle. Journal ofApplied Physiology 90, 1532–1538.

Series A, Biological Sciences and MedicalSciences 56, 240–247.

Roy, R., Monke, S., Allen, D. & Edgerton,V. (1999) Modulation of myonuclearnumber in functionally overloaded andexercised rat plantaris fibers. Journal ofApplied Physiology 87, 634–642.

Ruizeveld-De-Winter, J.A., Trapman, J.,Vermey, M. et al. (1991) Androgenreceptor expression in human tissues: an immunohistochemical study. Journalof Histochemistry and Cytochemistry 39,927–936.

Salmons, S. (1992) Myotrophic effects of ananabolic steroid in rabbit limb muscles.Muscle & Nerve 15, 806–812.

Sar, M., Lubahn, D.B., French, F.S. & Wilson, E.M. (1990)Immunohistochemical localization ofthe androgen receptor in rat and humantissues. Endocrinology 127, 3180–3186.

Sassoon, D. & Kelley, D.B. (1986) Thesexually dimorphic larynx of Xenopuslaevis: development and androgenregulation. American Journal of Anatomy177, 457–472.

Schaub, M.C., Brunner. U.T., Von-Schulthess, C., Neidhart, M. &Baumann, H. (1989) Adaptation ofcontractile proteins in human heart andskeletal muscles. Biomedica BiochimicaActa 48, S306–S312.

Schiaffino, S. & Reggiani, C. (1996)Molecular diversity of myofibrillarproteins: gene regulation and functionalsignificance. Physiological Reviews 76,371–423.

Schiaffino, S., Bormioli, S.P. & Aloisi, M.(1976) The fate of newly formed satellitecells during compensatory musclehypertrophy. Virchows Archiv. B: CellPathology 21, 113–118.

Schultz, E. & McCormick, K.M. (1994)Skeletal muscle satellite cells. Reviews ofPhysiology, Biochemistry and Pharmacology123, 213–257.

Staron, R.S., Leonardi, M.J., Karapondo,D.L. et al. (1991) Strength and skeletalmuscle adaptations in heavy-resistance-trained women after detraining andretraining. Journal of Applied Physiology70, 631–640.

Takagi, H., King, G.L., Robinson, G.S.,Ferrara, N. & Aiello, L.P. (1996)Adenosine mediates hypoxic inductionof vascular endothelial growth factor inretinal pericytes and endothelial cells.Investigative Ophthalmology and VisualScience 37, 2165–2176.

Takeda, H., Chodak, G., Mutchnik, S.,Nakamoto, T. & Chang, C. (1990)Immunohistochemical localization of

Pette, D. & Staron, R.S. (1997) Mammalianskeletal muscle fiber type transitions.International Review of Cytology 170,143–223.

Phillips, S.M., Tipton, K.D., Aarsland, A., Wolf, S.E. & Wolfe, R.R. (1997) Mixed muscle protein synthesis andbreakdown after resistance exercise inhumans. American Journal of Physiology273, E99–E107.

Powers, M.L. & Florini, J.R. (1975) A directeffect of testosterone on muscle cells in tissue culture. Endocrinology 97,1043–1047.

Quinn, L.S., Steinmetz, B., Maas, A., Ong, L. & Kaleko, M. (1994) Type-1insulin-like growth factor receptoroverexpression produces dual effects on myoblast proliferation anddifferentiation. Journal of CellularPhysiology 159, 387–398.

Regnier, M. & Herrera, A.A. (1993a)Changes in contractile properties by androgen hormones in sexuallydimorphic muscles of male frogs(Xenopus laevis). Journal of Physiology 461,565–581.

Regnier, M. & Herrera, A.A. (1993b)Differential sensitivity to androgenswithin a sexually dimorphic muscle ofmale frogs (Xenopus laevis). Journal ofNeurobiology 24, 1215–1228.

Richardson, R.S., Wagner, H., Mudaliar,S.R. et al. (1999) Human VEGF geneexpression in skeletal muscle: effect ofacute normoxic and hypoxic exercise.American Journal of Physiology 277,2247–2252.

Richardson, R.S., Wagner, H., Mudaliar,S.R. et al. (2000) Exercise adaptationattenuates VEGF gene expression inhuman skeletal muscle. American Journalof Physiology 279, 772–778.

Rogozkin, V. (1979) Metabolic effects ofanabolic steroid on skeletal muscle.Medicine and Science in Sports 11,160–163.

Rosenblatt, J.D. & Parry, D.J. (1992)Gamma irradiation preventscompensatory hypertrophy ofoverloaded mouse extensor digitorumlongus muscle. Journal of AppliedPhysiology 73, 2538–2543.

Rosenblatt, J.D., Yong, D. & Parry, D.J.(1994) Satellite cell activity is requiredfor hypertrophy of overloaded adult rat muscle. Muscle and Nerve 17, 608–613.

Roth, S.M., Martel, G.F., Ivey, F.M. et al.(2001) Skeletal muscle satellite cellcharacteristics in young and older menand women after heavy resistancestrength training. Journals of Gerontology.

Page 334: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

318 chapter 22

androgen receptors with mono- andpolyclonal antibodies to androgenreceptor. Journal of Endocrinology 126,17–25.

Tsika, R.W., Herrick, R.E. & Baldwin, K.M.(1987) Effect of anabolic steroids onskeletal muscle mass during hindlimbsuspension. Journal of Applied Physiology63, 2122–2127.

Wada, H., Ivester, C., Carabello, B.,Cooper, G. & Mcdermott, P. (1996)Translational initiation factor eIF-4E, alink between cardiac load and proteinsynthesis. Journal of Biological Chemistry271, 8359–8364.

Wagner, P.D., Masanes, F., Wagner, H. et al. (2001) Muscle angiogenic growthfactor gene responses to exercise inchronic renal failure. American Journal of Physiology 281, 539–546.

Wang, N., Hikida, R.S., Staron, R.S. &Simoneau, J.A. (1993) Muscle fiber types

Yamada, S., Buffinger, N., Dimario, J. &Strohman, R.C. (1989) Fibroblast growthfactor is stored in fiber extracellularmatrix and plays a role in regulatingmuscle hypertrophy. Medicine andScience in Sports and Exercise 21,S173–S180.

Yamaguchi, T.P. & Rossant, J. (1995)Fibroblast growth factors in mammaliandevelopment. Current Opinion in Geneticsand Development 5, 485–491.

Yan, Z., Biggs, R.B. & Booth, F.W. (1993) Insulin-like growth factorimmunoreactivity increases in muscleafter acute eccentric contractions. Journalof Applied Physiology 74, 410–414.

Yang, S., Alnaqeeb, M., Simpson, H. &Goldspink, G. (1996) Cloning andcharacterization of an IGF-1 isoformexpressed in skeletal muscle subjected tostretch. Journal of Muscle Research and CellMotility 17, 487–495.

of women after resistance training-quantitative ultrastructure and enzymeactivity. Pflugers Archiv 424, 494–502.

Willis, P.E., Chadan, S., Baracos, V. &Parkhouse, W.S. (1997) Acute exerciseattenuates age-associated resistance toinsulin-like growth factor I. AmericanJournal of Physiology 272, 397–404.

Willis, P.E., Chadan, S.G., Baracos, V. &Parkhouse, W.S. (1998) Restoration ofinsulin-like growth factor I action inskeletal muscle of old mice. AmericanJournal of Physiology 275, 525–530.

Winchester, P.K. & Gonyea, W.J. (1992) Aquantitative study of satellite cells andmyonuclei in stretched avian slow tonicmuscle. Anatomical Record 232, 369–377.

Wong, T.S. & Booth, F.W. (1990) Proteinmetabolism in rat gastrocnemius muscleafter stimulated chronic concentricexercise. Journal of Applied Physiology 69,1709–1717.

Page 335: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

319

The male sex hormone, testosterone, influences notonly sexual activity and emotional behavior (e.g.aggressiveness) but also contributes to metaboliccontrol. Testosterone is considered to be an anabolichormone; however, its role in metabolic control isactually more extended. Testosterone influences byits contra-action effects on several other hormones.In resistance training, the main role of testosteroneis the induction of synthesis of contractile pro-teins in involved muscles. Beside that, during acuteresistance exercises, as well as during competition,testosterone action seems to be essential for mobiliz-ing performance capacity.

The pituitary–testicular system

Testosterone is produced by the interstitial cells ofLeydig, constituting of 20% of the mass of testes inadult men. Quantitatively, a much lesser amount oftestosterone is derivated from androgenic steroidsformed in the adrenal cortex. The Leydig cellssecrete testosterone only when they are stimulatedby lutropin (LH; also called luteinizing hormone)from the pituitary gland. The quantity of secretedtestosterone is in direct proportion to the amount of LH available. The secretion of LH is stimulated by a hypothalamic neurohormone, gonadotropin-releasing hormone (GnRH). In turn, blood testo-sterone inhibits GnRH secretion (a pronounced negative feedback effect) as well as LH secretion bythe pituitary (a weak negative feedback effect). Toolittle testosterone allows the neurosecretory cells ofthalamus to secrete large amounts of GnRH withfollowed increases in secretion of pituitary LH and

testicular testosterone. Thus, the pituitary gonado-graphs secreting LH and the testes constitute thepituitary–testicular system, and its activity is con-trolled by nervous influences reaching the pituitarygland from the hypothalamus via neurosecretionand by the feedback action of blood testosterone.

Both LH-releasing hormone and LH are secretedin an episodic manner. Secretory bursts of GnRHlast a few minutes at a time, once every 1–3 h. Thepulsative manner of testosterone is less pronounced.The blood testosterone level is highest early in themorning and decreases during the day. Most of the testosterone in blood is bound by sex hormonebinding globulin (SHBG).

Metabolic action of testosterone

Anabolic action. In 1935 Kochakian reported that, inmale castrated dogs, testosterone injection resultedin long-lasting nitrogen retention (Kochakian 1935).Over the next few years, several papers confirmedthis result and showed that castration of male rats,guinea pigs or mice induces weakened skeletalmuscles due to a decreased protein synthesis rate.Substitution therapy with testosterone abolishedthese castration effects (Papanicolaou & Falk 1938;Scow & Hayes 1955; Kochakian 1959; Kochakian et al. 1964).

According to present evidence, metabolic actionof testosterone is mediated through the androgenreceptor in the cytoplasm of cell. In the cell cyto-plasm testosterone is bound by the receptor protein.The formed complex is activated and transferredinto the cell nucleus. Binding of the complex by

Chapter 23

Resistance Exercise and Testosterone

ATKO VIRU AND MEHIS VIRU

Page 336: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

320 chapter 23

chromosomal proteins triggers the production ofmRNA specific for the synthesis of necessary pro-tein(s) responsible for actualization of the testo-sterone effect. In the skeletal muscle the main locus of testosterone action is the synthesis of myofibril-lar proteins. This way testosterone contributes to the development of muscular hypertrophy. Thereceptor protein is common for testosterone andseveral other androgens. In skeletal muscle fibers, as well as in bone cells, testosterone has a higheraffinity to receptor than other androgens. In othercells the highest affinity belongs to 5α-dihydro-testosterone. In these cases, after testosterone entersthe cell it has to be converted to 5α-dihydrotesto-sterone. Conversion of testosterone to dihydrotesto-sterone is not found in muscle fibers and bone cells.The information contained in the mRNA is trans-lated for the synthesis of related cellular proteins inribosomes.

The state of cellular receptors is regulated byincreasing/decreasing the number of binding sitesand by changing the binding affinity. Thus, testoster-one metabolism depends on testosterone produc-tion as well as the up- or down-regulation of cellularspecific receptor. Therefore, the metabolic effect ofthe hormone augments not only by increased con-centration of the hormone in the intercellular fluidbut also as a result of the increased number of bind-ing sites and/or increased affinity of receptor to hor-mone. In conditions of receptor down-regulation,the high hormone level cannot produce a pronouncedmetabolic effect. Consequently, it is incorrect to putthe sign of equality between the blood concentrationof a hormone and its metabolic effect.

Ontogenetic development of skeletal muscle is deeplyrelated to the metabolic effect of testosterone caus-ing the male body build to develop from the finalstages of puberty, and it is characterized by theincrease of musculature over that of the female.Increased testosterone level after sexual maturationalso warrants good faculties for strength, power andspeed training.

Anti-catabolic effects. Testosterone is capable ofinhibiting the catabolic effect of glucocortioids (anti-catabolic action) as well as reducing the suppression

of protein synthesis (anti-anabolic action) exerted by them. Anti-catabolic action and the inhibition ofanti-anabolic action are founded on the competitionbetween testosterone and cortisol for the specificcellular receptors of glucocorticoids. Depending onthe amount of glucocorticoid receptors occupied bytestosterone, the cortisol catabolic and anti-anabolicactions decrease (Mayer & Rosen 1977).

Stimulation of bone growth and calcium retention.Testosterone increases the total bone matrix andcauses calcium retention. After puberty, bones growconsiderably in thickness and deposit additionalamounts of calcium salts (Ritzen et al. 1981; Krabbeet al. 1982). The increase in bone matrix has beenrelated to the anabolic function of testosterone, andthe deposition of calcium salts is secondarily causedby the increase of bone matrix.

Other effects. During adolescence and early adult-hood, testosterone increases the basal metabolism5–10%. Testosterone is capable of increasing the rateof erythropoiesis (Palacios et al. 1983). It also exerts amodest influence on sodium reabsorption in renaltubules.

Testosterone in women

Testosterone in female blood plasma originatesmainly from the adrenal cortex as a byproduct of glu-cocorticoid biosynthesis. The secretion of adrenalcortex contains androgenic steroids that can beperipherally converted to testosterone. Adrenalsbegin to produce androgenic steroids at the begin-ning of the second decade of postnatal life in rela-tion to the adrenarche. The marker of this process isthe serum level of dehydroepiandrosterone sulfate(DHEAS) (Wierman & Crowley 1986).

The production of testosterone in women dependson the rate of biosynthesis of glucocorticoids stimu-lated by adrenocorticotropic hormone (ACTH) fromthe anterior lobe of pituitary gland. Thus, in womenLH has only a minor role, if at all, in the control ofthe blood level of testosterone. Therefore, whencomparing various responses of blood testosteronein men and women, the principal gender differencein the control mechanism has to borne in the mind.

Page 337: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 321

Although the blood level of testosterone in adultwomen is about 10 times lower than in men, themetabolic effects of testosterone are not so less pronounced. In women, testosterone metaboliceffects increase in relation to estrogen production(Danhaive & Rousseau 1988). It has been suggestedthat increased sensitivity in regard to testosteronemetabolic effect is related to up-regulation of andro-genic receptor. Another possibility is that in womentestosterone has favored conditions for competitionfor glucocorticoid receptors.

Testosterone responses in resistanceexercise

Exercise-induced hormonal responses depend onfour main determinants: (i) exercise intensity; (ii)exercise duration; (iii) level of adaptation to the concrete form of exercise; and (iv) homeostaticneeds (Viru, A. 1992; Viru, A. et al. 1996). The actionof these determinants is modified by several modu-lators, such as emotional strain, carbohydrate andoxygen availability, environmental temperature,biorhythms and fatigue (Viru, A. et al. 1996).

Three mechanisms are assumed to constitute thelink between exercise and endocrine activities. Oneof them is related to triggering nervous dischargefrom cerebral motor centers to spinal motoneurons(central motor command). The importance of thisfor the activation of endocrine function has beenshown in experiments that injected tubocurarine in men. A bolus of 0.015 mg·kg–1 α-tubocurarinecaused a partial peripheral neuromuscular block-ade. As a result, it weakened the skeletal muscles.Therefore, a stronger voluntary effort was necessaryto produce a certain work output compared withnormal conditions. The increased voluntary effortwas confirmed by the higher rate of perceived exer-tion in this experiment (Galbo et al. 1987). The‘stronger’ central motor command was associatedwith exaggerated catecholamine, growth hormoneand ACTH responses during exercise performed ata similar level of oxygen uptake without neuromus-cular blockade (Kjær et al. 1987).

Galbo (1983) assumed that, during continuousexercises, hormone responses were influenced byimpulses from receptors sensing temperature, intra-

vascular volume, oxygen tension and glucose avail-ability. However, attention should have first beengiven to the feedback by nervous impulses fromreceptors located in skeletal muscles (propriocep-tors sensing the muscular tension and metaborecep-tors reacting to metabolite accumulation). The useof small doses of epidural anesthesia, to block thethin sensory afferents (mostly from metaborecep-tors) and leave almost intact the thicker efferentfibers and, subsequently, motor function, allowedthe authors to demonstrate the essential role of nervous feedback from muscles for ACTH and β-endorphin response (Kjær et al. 1989).

Significance of exercise intensity, duration andrest intervals for testosterone responses inresistance exercises

In cyclic exercises the level of intensity has beenfound to be significant for several hormone res-ponses. Exercise intensities higher than the cor-responding threshold cause hormonal responses.Intensity thresholds for catecholamines (Lehmannet al. 1991), ACTH (Rahkila et al. 1988), cortisol (Port1981), β-endorphin (Rahkila et al. 1988) and growthhormone (Chwalbinska-Moneta et al. 1996) are closeto the anaerobic threshold. In men performing acyclic incremental exercise test, the exercise intensityhas been noticed above that of the elevated bloodlevel of testosterone (Jezova et al. 1985). The samewas indicated by the results of Galbo et al. (1977).However, results of several other studies failed toindicate a clear-cut dependence on exercise intensity.

Taking into the consideration the significance of the ‘strength’ of the central motor command,most resistance and power exercises should be in-tensive enough to evoke sufficiently strong centralmotor command for triggering hormone responses.Nevertheless, an instantaneous single application ofgreat force or muscle power does not mostly inducehormonal responses. The duration of the effort orthe number of repetitions seems to be important aswell. Kraemer, W.J. et al. (1989, 1991b) compared the action of cycling when 100%, 73%, 55% or 36% of leg power was applied for pedaling. The max-imal duration of exercise was 6 s at 100%, 10 s at 73%,47 s at 55% and 3 min 31 s at 36%. In the case of the

Page 338: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

322 chapter 23

highest exercise intensity, the 6 s of duration was suf-ficient to trigger the increase of norepinephrine inblood serum but not to elevate levels of epinephrineand cortisol. These results demonstrate that, in exer-cises of high power output, the triggering of hor-monal responses depends on the combined effect ofthe rate of power output and duration of exercise.

Kinetics of testosterone response. The rate of actualappearance of hormonal response is related to: (i)the rate of reaching the triggering signal to the endo-crine gland (the highest rate possessed to nervousactivation of adrenal medulla; a comparatively highrate is signaling the activation of anterior lobe of the pituitary gland with the aid of hypothalamusneurohormones); (ii) the possibilities for immediatesecretion of the already biosynthesized hormone(e.g. catecholamines bound in cytoplasmic granulesof adrenomedullary cells); (iii) the rate of biosyn-thesis of hormone in quantity necessary for increas-ing the blood level of hormone and maintaining that level during the exercise. The kinetics of testo-sterone production has still not been investigated inthe exercise situation. Padron et al. (1980) indicatedthat a single injection of human chorinic gonadi-tropin in men was followed 2 h later by a prolongedincrease of testosterone level in the blood. Theseresults suggest that the rate of secretory response oftestosterone depends on the rate of synthesis of thishormone. Obviously, we have to assume that tes-tosterone secretion cannot be enhanced rapidly.Variability of testosterone changes in exercise ofshort duration rather shows the normal fluctuationsof the basal level of testosterone than the actualsecretory response.

One-minute duration of high intensity exercise(consecutive vertical jumps repeated at maximalrate) evoked increases of concentration of anteriorpituitary and thyroid hormones 20–36%, but nochange of growth hormone, prolactin and insulin-like growth factor concentrations. Total and freetestosterone and cortisol level elevated only 12–14%(Bosco et al. 1996a). The magnitude of changes oftestosterone and cortisol was similar to the reduc-tion of plasma volume in short-term highly intensecyclic exercises (Sejersted et al. 1986). Therefore, theobserved testosterone and cortisol changes were

related, obviously, to the hemoconcentration. Galboet al. (1977) interpreted the increase of testoster-one concentration by 13% in incremental exercise as a manifestation of plasma volume reduction.Kraemer, W.J. et al. (1992) also pointed to the pos-sibility that in moderate resistance exercise sessionsthe elevated testosterone levels may be related tochanges in plasma volume. Wilkerson et al. (1980)measured simultaneously changes of plasma vol-ume and testosterone concentration during cyclicexercise. Their conclusion was that in 20-min steadystate exercises the modest increase of testosteroneconcentration was due to reduction of plasma vol-ume but not due to elevated testosterone secretion.An additional reason for testosterone increase inblood without elevated secretion is the reducedmetabolic clearance rate of testosterone during exer-cise (Sutton et al. 1978).

According to these results, the actual increase oftestosterone secretion in exercise should be a delayedresponse. Nevertheless, Jezova et al. (1985) found asignificant increase of testosterone concentration(by 28%) after cycloergometric testing of 4.5 minduration performed at very high intensity (5 W·kgbody weight–1). Obviously, the exercises cause arather complicated situation including the possibil-ity for altered kinetics of testosterone response.

Results of Ahtiainen et al. (2003) showed that aftertwo sets of leg press at 12-repetition maximum (RM)(rest between sets 2 min) free and total testosteroneconcentrations were significantly higher than initialvalues. The increased levels persisted up to the endof session, whereas a trend for further elevation offree hormone level was observed (Fig. 23.1). Sinceplasma volume decreased by 11.8% during the session in one protocol (maximum repetition) andby 14.4% in the other protocol (‘forced’ repetition),free testosterone response surpassed the hemo-concentration effect after first two sets and totaltestosterone response after six to eight sets. Anotherstudy of this research team demonstrated that foursets of squats of 10-RM (rest pauses 90 s) resulted in increases of concentrations of free and totaltestosterone as well as of ACTH, cortisol and lactate(Kraemer, W.J. et al. 1998a). However, again the con-tribution of the hemoconcentration effect remains.All in all it is necessary to bear in mind that the

Page 339: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 323

responses of the blood testosterone concentra-tion do not linearly reflect the secretion responses.Nevertheless, concentration changes are importantbecause metabolic effects of hormones are related tohormone concentration in intercellular fluid. Thetotal hormone amount in the body fluid compart-ments has significance for maintaining the hormoneeffects.

Significance of intensity and duration of resistance ses-sion. After sessions of resistance exercises a frequentfinding is increased testosterone levels. This resulthas been obtained in results of sessions with a dura-tion of approximately 30 min (Jürimäe et al. 1990), orafter four exercises consisting of either three to five

sets of 5-RM over 3-min rest pauses or three sets of10-RM over 1-min rest pauses (Kraemer, W.J. et al.1990, 1991a). However, there is also another pos-sibility: six series of eight bench presses at 70% ofmaximal resistance (total 24 min) did not cause asignificant increase of testosterone level. Testosteroneresponse was also not observed when the sessionwas followed by a maximal number of consecutivebench presses at 70% (Guezennec et al. 1986).

When two workload protocols (10-RM vs. 5-RM)were involved, testosterone increase in blood wasgreater after the four exercises using 10-RM (i.e.lower weight, greater total work) protocol as com-pared with 5-RM (i.e. higher weight, lower totalwork). After eight exercises no significant difference

Perc

ent

40

35

30

25

20

15

10

5

0

Free testosterone

Pre 2 sets 4 sets 2 sets 2 sets Post-15 min(a)

Post-30 min

Perc

ent

20

15

10

5

0

–5

–10

–15

–20

–25

Testosterone

Pre 2 sets 4 sets 2 sets 2 sets Post-15 min

(b)

Post-30 min

Fig. 23.1 Dynamics of testosteroneduring a resistance training session.(a) Dynamics of free testosterone. (b) Dynamics of total testosterone.Interrupted horizontal line,magnitude of hemoconcentration.(Data from Ahtiainen et al. 2003.)

Page 340: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

324 chapter 23

in testosterone levels was found between the twoprotocols (Kraemer, W.J. et al. 1991a). Volvek et al.(1997a) observed that testosterone concentrationsincreased modestly (by 7%) using a bench press protocol of five sets to failure with 10-RM butsignificantly more (by 15%) using a jump squat pro-tocol of 15 sets with 10 repetitions at 30% of 1-RMsquat. The significance of training session workloadwas confirmed by Cotschalk et al. (1997): a three-setheavy resistance protocol resulted in a greater in-crease in testosterone concentration than a one-setprotocol. Häkkinen and Pakarinen (1993) reportedthat the increase of the total workload is responsiblefor the blood testosterone response despite the neces-sary reduction of the exercise intensity.

Besides the characteristics of exercise, duration ofrest intervals has a significant effect on hormonalresponses. The significance of rest intervals un-doubtably influenced the results of above-mentionedstudies of Kraemer, W.J. et al. (1990, 1991a). After a 30-min intensive single circuit weight training ses-sion (work/rest ratio 30 s; 30 s at 70% of 1-RM) maleuniversity students exhibited testosterone levels24% higher than the initial values (Jürimäe et al.1990).

Summing up, Fleck and Kraemer (1997) affirmedthat the hormonal response in resistance exercisedepends on muscle mass recruited, intensity ofworkout, amount of rest between sets and exercisesand previous training. Both concentric and eccentricexercises are able to increase the testosterone con-centration in blood (Durand et al. 2003).

Jensen et al. (1991) compared testosterone changesduring and after endurance and strength trainingsessions in the same men for both forms of training.The mean increase of testosterone concentrationwas 27% in the endurance session and 37% in thestrength session. Differences between responses inthe two sessions were not significant but there werelarge differences in the testosterone responses at theindividual level. A high correlation (r = 0.98) forindividuals was found between increases of testo-sterone concentration after strength and endur-ance sessions. The authors suggested that the interindividual differences in testosterone responsesmight be of importance for individual adaptation totraining.

Power sessions. A large number of repetitions with low power output in bodybuilders decreasedtestosterone and increased growth hormone levels,whereas in weightlifters a high number of repeti-tions increased testosterone concentrations withoutchanges in growth hormone level. When power out-put remained close to maximum and the applicationof force increased (and number of series decreased),no significant hormone changes were found inweightlifters. Sprinters performed exercises at max-imal power with a force of 60%. Although the num-ber of series was modest, the total workload wasobviously high. In men, blood concentration of tes-tosterone decreased together with the same changein blood levels of LH and cortisol. It is possible tosuggest that the reversed hormonal responses wererelated to pronounced fatigue that developed dur-ing the session. This possibility was confirmed by asignificant decrease of average power in full squatsand by an increase of the total bioelectric activity ofcontracting muscles to power output in full squats(Bosco et al. 2000).

Training and modulators effects

Training effects. Few studies provide results on training effects in resistance training sessions.Guezennec et al. (1986) observed no significanttestosterone response after six series of eight benchpresses at 70% of 1-RM or after the maximal number of repetitions at the same workload. The measure-ments were repeated each month for 4 months oftraining, but the results were the same. Because thesubjects were male weight-trained athletes, the lackof testosterone response might be related to previ-ous training adaptation to test exercise. In studies onelite athletes, Häkkinen and Pakarinen (1993) foundthat 20 sets of squats at 1-RM did not increase con-centrations of total and free testosterone, whereasthe testosterone level rose significantly when 10 setswere performed at 70% of 10-RM. If these results are related to previous adaptation to resistance exercises then, interestingly, adaptation influencesthe response to high intensity workload but not tovoluminous workload.

In weightlifters, increased testosterone concentra-tion has been found after four series of six squats at

Page 341: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 325

90–95% of 6-RM as well as after nine to 10 squats at60–65% of 6-RM (Schwab et al. 1993). In these ath-letes the exercises did not demonstrate adaptationby disappearance of testosterone responses. How-ever, since the test exercise was adjusted to the individual repetition maximum, the increased actualworkload avoided the disappearance of testoster-one response.

Summing up, resistance training may remove thehormonal responses when the test exercise is similarto those frequently used in training. At the sametime, a possibility exists that long-term resistancetraining promotes testosterone responses, at least inadolescent athletes. The same weightlifting protocolincreased testosterone concentration in 17-year-oldjuniors who had more than 2-years training experi-ence (Kraemer, W.J. et al. 1992).

The effects of resistance training differ from theoutcome of endurance training, not only by changesat the level of muscle fibers and of aerobic capacity,but also in regard to influence on hormonal res-ponses in exercises. The combination of strengthand endurance training results in an attenuation ofthe performance improvements and adaptationstypical for single-mode training (Kraemer, W.J. et al. 1995a). Endurance training suppresses bloodtestosterone level (Hackney 1996). The endogenousstimulation of testosterone production gives lesspronounced change in endurance-trained men com-pared to sedentary men (Hackney et al. 2003).

Modulator influences on testosterone responses in resist-ance exercise. It has been believed that aggressivenessfavors the performance of resistance and power exer-cises. At the same time, aggressiveness is related totestosterone. Therefore a question arises as to whetheremotions of this type influence testosterone res-ponses in resistance exercises. In regard to this ques-tion, a new way of thinking arose from the results ofElias (1981). This study showed that judo fightingincreased the blood level of testosterone and cortisolmore in the winners than in the losers. In accord-ance, tennis players who exhibited a good mood andself-assurance had higher testosterone levels beforecompetition. After the match, winners exhibitedfurther elevation of the testosterone level, a decreasebeing observed in losers (Booth, A. et al. 1989).

Subjects with a high trait anxiety showed signific-ant increases of testosterone in test exercise similarto normal subjects, whereas concentrations of anadrenal androgen, androstendione, increased less in subjects displaying a high trait anxiety (Diamondet al. 1989).

The above results presented that after a hardpower training session a pronounced decrease wasfound in concentrations of testosterone and cortisol(Bosco et al. 2000). It was suggested that this changewas related to fatigue; more correctly, to a latentfatigue appearing before the actual drop of workingcapacity. This suggestion has to be verified with theaid of further experiments.

Häkkinen et al. (1988b) measured hormone res-ponses in elite athletes performing two strengthtraining sessions a day. Both testosterone and cor-tisol responses decreased after the first session, butthe testosterone increased after the second session.The authors explained these results by the circadianbiorhythm: during the morning session the testo-sterone increase was masked by the decline in hor-mone basal level, during the afternoon session thelowered initial level of testosterone favored itsincrease.

Results also became available showing that diet-ary nutrients (Volek et al. 1997b), a protein–carbohy-drate supplement consumed 2 h before (Kraemer,W.J. et al. 1998b) or post-exercise nutrition (Bloomeret al. 2000) may influence the testosterone pattern in resistance training session or during the 24 h ofrecovery. Amino acid supplementation (Fry et al.1993) or the administration of either creatine (Voleket al. 1997a; Op’Teijnde & Hespel 2001) or ginseng(Youl et al. 2002) failed to alter testosteroneresponses.

Relationship of testosterone increase to LH response.Some studies indicated concomitant increases oftestosterone and LH levels during long-lastingtraining (Häkkinen et al. 1987, 1988a). However, apossibility of uncorrelated changes of these two hor-mones has also been shown (Häkkinen & Pakarinen1991). In resistance exercise sessions increases oftestosterone and LH are usually not parallel. Insome studies concomitant declines of testosteroneand LH have been observed (Häkkinen et al. 1988b;

Page 342: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

326 chapter 23

Bosco et al. 2000). However, these data cannot beused as evidence against the role of LH in testo-sterone response during exercises. The LH responseconsists of a short-term burst of secretion, which isfollowed by a delayed increase in testosterone secre-tion. Therefore, in experiments it is a complicatedtask to ‘catch’ the LH response. More convenient isto record the prolonged result of LH pulsation inincreased testosterone secretion and blood level.The exercise effect on pulsative secretion of LH hasbeen studied in women and in relation to enduranceexercises (Cumming et al. 1985; Weltman et al. 1990)but not in men and in regard to resistance exercise.Endurance training may alter the biological activityof endogenous LH (Di Luigi et al. 2002). Corres-ponding information regarding resistance trainingis not available.

Gender and age

Testosterone responses in women. Several studies indicated that, differently from men, women do notreact to resistance exercises by increased testos-terone level in blood. When eight exercises for various muscles were performed with workloads of either 5-RM (rest intervals 3 min) or 10-RM (rest intervals 1 min), testosterone concentrationincreased in men but not in women despite priorrecreational resistance training experience in bothcontingents (Kraemer, W.J. et al. 1991a, 1993). In con-trast, Cumming et al. (1987) found a 20% increase oftestosterone in women after a session consisting ofsix isokinetic resistance exercises. Although the LHlevel increased by 60%, it is unlikely that it was thephysiological stimulus for testosterone productionbecause women produce this hormone as a bypro-duct of steroid biosynthesis in the adrenal cortex.Accordingly, those women who did not exhibit acortisol response failed to show an increase in tes-tosterone concentrations. The other female subjectsshowed a parallel increase in testosterone and cor-tisol concentrations (Cumming et al. 1987).

During strength exercises, Weiss et al. (1983)observed rather similar relative increases in bloodtestosterone, in men from the high level by 21.6%and in women from the low level by 16.7%. Andro-stendione concentration increased significantly in

both gender groups without difference in the mag-nitude of response. Resistance exercises during 40 min increased concentration of another adrenalandrogen, DHEA, more than endurance exercises inwomen 19–69 years of age (Copeland et al. 2002).

Kraemer, R.R. et al. (1995) compared hormonalresponses in healthy women in early follicular andluteal phases. A low-volume resistance exerciseprogram caused greater estradiol response in thefollicular phase. Growth hormone and andros-tendione responses appeared only in luteal phase,whereas testosterone and progesterone did notresponse during this exercise.

A problem is whether the resistance exercisesinfluence the activity of enzymes in the adrenal cortex responsible for forming androgen steroids.Neither confirming nor contradicting evidence isstill available. However, individual differences inthe production of androgens exist between women.It has been shown that an acute resistance exercisetest (ARET) increased concentrations of total testo-sterone by 25%, free testosterone by 25% and SHBGby 4%. Multiple regression analysis indicated that inyoung healthy women the testosterone responses to ARET is predicted by the subscapular to tricepsratio in skinfolds and by the ratio of upper-arm fatto mid-thigh fat assessed with magnetic resonanceimaging. Waist-to-hip ratio in fat failed to have sig-nificance as a discriminator for hormonal concen-trations in women (Nindl et al. 2001).

Testosterone responses in elderly people. According tothe results of Häkkinen and Pakarinenen (1995), a heavy resistance session caused testosterone in-crease in both 30-year-old and 50-year-old men, butnot in 70-year-old men. Women of any age failed toshow testosterone response. In another study,Häkkinen et al. (1998) compared the acute effect of heavy resistance exercise on blood testosterone in young (mean age 26.5 years) and old (mean age 70 years) men. Exercises for legs, upper body andtheir combination increased total testosterone levelin young men but only leg exercise increased totaltestosterone level in old men. Kraemer, W.J. et al.(1998a) found that four sets of squats of 10-RMcaused increased total and free testosterone levels inboth 30- and 62-year-old men. The area under the

Page 343: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 327

response curve during and 30-min post-exercisewas greater in younger men.

Testosterone responses in adolescents. Appearance oftestosterone in blood is related to puberty in boysand to adrenarche in girls. During cyclic exercisessignificant increase of testosterone level has beendetected from the stage 4 of sexual maturation (byTanner’s scale) in boys (Fahey et al. 1979) and fromthe stage 5 in girls (Viru, A. et al. 1998).

In resistance exercises, post-exercise as well aspre-exercise testosterone levels were lower in 15-year-old male athletes than in adult male athletes(Pullinen et al. 1998).

Pattern of blood testosterone during the recovery period

In men, the testosterone level gradually returns toits initial level during the 1st hour following resist-ance sessions. In females, low blood levels of testo-sterone remain without significant alterations bothduring the session and the 1st post-exercise hour(Kraemer, W.J. et al. 1991a). This pattern is differentfrom that found after cyclic exercises. After 2 h ofbicycle ergometric exercise the testosterone leveldropped during the 6 h post-exercise. Twenty-fourhours later it showed a normalizing trend, but themean testosterone concentration was still below theinitial values both in trained and untrained youngmen (Viru, A. et al. 1992). Similar results have beenobtained by a Finnish research team (Kuoppasalmi1980; Kuoppasalmi et al. 1980): after running for13–14 km the testosterone level dropped during the1st hour and then remained low for 5 h. Ahtiainen et al. (2003) also found that 30 min after a resistancetraining session, free and total testosterone levelswere below initial levels. When a 30-min resist-ance session protocol prescribed a work-to-rest ratioof 30 s, 30-s testosterone concentration decreased during the first 6 h of the post-exercise recovery(Jürimäe et al. 1990). Thus, the testosterone patternafter resistance exercises may be similar to that afterendurance exercises. In the first and second morn-ing after heavy resistance training, the level of totaltestosterone was normal but the level of free testo-sterone increased (Ahtiainen et al. 2003).

In rats, seven times 1-min swimming with a highadditional weight (of 12% body mass) simulatedefforts in resistance training. The blood level oftestosterone increased slightly. During the first 2 hof restitution the hormone level decreased. Fourhours after exercise a secondary rise in the hormoneconcentration was detected, exceeding the restinglevel by 1.5–2.5 times. Testosterone levels in cyto-plasm of skeletal and heart muscle fibers increased,with the peak values 72 h after swimming (Fig.23.2). At that time the specific binding of andro-gens, as well as the number of binding sites, wereincreased in the cytoplasm of skeletal muscle fibers(Tchaikovski et al. 1986).

It is possible to ask what is important for the post-exercise induction of adaptive protein synthesis in exercised muscles, either testosterone responseduring the exercise or the secondary post-exercisetestosterone increase. The presented results of ratexperiments suggest that both increases of testo-sterone constitute an entire response.

Testosterone in monitoring of resistance training

The fact that something is measured in athletes doesnot mean training monitoring. It is possible to speakabout training monitoring if the following five prin-ciples are completely followed (Viru, A. & Viru 2001):1 It is a process performed for the purpose of in-creasing the effectiveness of training guidance.2 It is based on recording changes in an athlete during various stages of the training or under theinfluence of main elements of the sport activities.3 It is a highly specific process depending on thesport event, performance level of the athlete andage/gender peculiarities.4 Any method or measurement is in a sense train-ing monitoring if it provides reliable informationrelated to the task being monitored.5 The information obtained from measurementshas to be understandable so that necessary correct-ive changes in training can be made.In the monitoring of the resistance training testo-sterone basal level, responses to test exercise and theratio of testosterone/cortisol have been used. Theidea is to get information about the contribution

Page 344: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

328 chapter 23

of testosterone in training adaptations, and to beinformed about excessive training workloads.

Training effects

Testosterone basal level during prolonged resistancetraining. The related studies were initiated byHäkkinen from Finland. In 1985 he and his collabor-ators reported that heavy resistance training for 24weeks did not change the basal level of testosteronein blood. Blood cortisol level decreased; the testo-sterone/cortisol ratio increased. This change wasparallel to the increase in isometric force of the legextensor muscles. During the followed detrainingfor 12 weeks, both indices decreased to the pretrain-ing values. In combined training using jumping andstrengthened exercises (weights of 60–80% 1-RM),

during the first 8 weeks the testosterone levelincreased and the cortisol level decreased. After 16weeks of training, levels of both hormones decreased.At the end of 24th week the mean concentrations of testosterone and cortisol did not differ from theinitial values. The detraining did not accompanyhormonal changes. The effect of this training pro-tocol on isometric force was significantly less pro-nounced than that in the high-resistance traininggroup (Häkkinen et al. 1985).

Blood samples obtained every 4 months in eliteweightlifters showed testosterone increased fromthe month 8 to month 12 (Häkkinen et al. 1987).During the next year the increased testosteronelevel persisted (Häkkinen et al. 1988a). The authorssuggested that increased testosterone level mightcreate optimal conditions for intensive weight

pg

·mL–1

2400

2100

1800

1500

1200

900

600

300

0

pg

·g–1

600

500

400

300

200

100

0

Testosterone

Exercise(a) 0 h 2 h 4 h 12 h 24 h 48 h 72 h 144 h

SerumCytosol

cpm

1400

1200

1000

800

600

400

200

0

fmo

l·mg

pro

tein

–1

1.0

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

0.0Exercise(b) 0 h 2 h 4 h 12 h 24 h 48 h 72 h 144 h

Specific bindingNumber of binding sites

Fig. 23.2 Post-exercise dynamics of testosterone and its binding inskeletal muscle after swimming inrats. (a) Closed triangles, testosteronein serum. Open circles, testosteronein cytosol of muscle fibers. (b) Open squares, specific binding oftestosterone. Closed rhombs, numberof binding sites. (Data fromTschaikovski et al. 1986.)

Page 345: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 329

training to increase strength improvement in elite-strength athletes. In 1990 this team reported that,according to 51 weeks of follow-up of six eliteweightlifters, the fitness level (evaluated by arbit-rary units) and blood testosterone level changed incorrelation (Busso et al. 1990).

Effects of hard-training stages. Six weeks of pre-paratory training of elite weightlifters was dividedinto a high voluminous stressful stage for 2 weeksfollowed by 2 weeks of ‘normal’ training and then 2weeks of taper. During the first 2 weeks testoster-one concentration decreased significantly, cortisolconcentration elevated slightly and the ratio of testosterone/cortisol dropped. During next 2 weekschanges were modest, if at all. (Häkkinen et al. 1987).

The results of Busso et al. (1992) confirmed that in elite weightlifters a 4-week stage of intensive train-ing was accompanied with decreases in the bloodtestosterone level. Fry et al. (1994) investigated 1week of intensive training causing overreaching injunior weightlifters. During the 1st year the intensetraining week resulted in attenuated testosteroneresponses in test exercise. Over the 2nd year theintensive training augmented the testosteroneresponses.

Kraemer, W.J. et al. (1995a) compared the effectsof hard endurance and strength training and theircombination. During endurance training, testoster-one was constant but undulations occurred in thecortisol response. In strength training, testosteroneagain stayed constant while cortisol response de-creased. Combined training demonstrated changesof both testosterone and cortisol levels over thetraining period.

In endurance athletes hard-training stages resultin two phases of hormonal changes (Viru, A. & Viru2001). Typical for the 1st phase was increased corti-sol basal level and exaggerated cortisol increase instrenuous test exercise. Testosterone basal level wasconstant or reduced and responses in test exerciseattenuated. The 2nd phase was indicated by sup-pressed cortisol response in exercise while the basal level might be either increased or decreased.Testosterone levels before and after test exercisewere mostly low. Although the material is far fromsufficient for conclusions, two phases may be sug-

gested in resistance training under the influence ofincreased training volume. However, available datasuggests that in strength athletes most typical arechanges not in the cortisol level but in the testo-sterone level.

The meaning of ‘hard-training stage’ is a relativeone depending on previous training. Therefore, thelack of testosterone changes in ‘intense’ training fora couple of weeks (Häkkinen & Pakarinen 1991) or 8 weeks of ‘heavy’ resistance training (Hickson et al.1994; Potteinger et al. 1995) does not contradict theresults of other studies. Also 12 weeks of heavyresistance training did not change either the restinglevel of testosterone or the responses in training sessions (McCall et al. 1999).

Kraemer, W.J. et al. (1996) suggests that in heavytraining hormonal changes are related to earlyadaptation, which later disappears. A questionarises in whether the decrease of hormonal changesindicates the necessity for further increase in thetraining workout.

Testosterone in the taper stage. During the taper stageafter ‘stressful’ training (causing a decrease in tes-tosterone and a slight increase in cortisol levels),testosterone remained low and cortisol decreased(Häkkinen et al. 1987). However, different situationsmay exist. From the data of Busso et al. (1990) it ispossible that a sharp reduction of training workloadassociates with the lowest testosterone level duringthe training year. Another report of this team (Bussoet al. 1992) also showed that the reduction of trainingworkload for 2 weeks did not stop the testosteronedecline that appeared during the previous 4 weeksof hard training. In power athletes a 2-week detrain-ing after heavy resistance training caused a decreaseof cortisol and an increase of testosterone concentra-tion. Detraining does not impair the neuromuscularperformance, as indicated by the results of mostmuscle strength and power tests and surface elec-tromyogram (EMG) activity (Hortobagyi et al. 1993).In recreationally strength-trained men, 6 weeks ofdetraining produced only a minimal change in per-formance and hormonal levels (Kraemer, W.J. et al.2002).

The taper problem is a rather complicated onebecause the taper stage may consist of various

Page 346: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

330 chapter 23

degrees of reduction in the training load and in various designs in regard to the choice of exercisesand the pattern of their intensity. Further, the bodystate when the reduction of training is considered to be necessary may be also different. Moreover, the body responses both to overtraining and taperare individually variable. Therefore, summing-upresults obtained in various athletes may actuallycause a loss of important information.

Discussion of hormonal changes in the taper stageled to an assumption that before competition thetaper stage has to be substituted by a short-termtraining stage for preconditioning the peak (at leasthigh) performance. For power and strength events,a high testosterone basal level may constitute animportant precondition for successful performance(Viru, M. & Viru 2000). This is a problem still wait-ing systematic research.

Overtraining. The main hormonal indicators of overtraining are suppressed production of cortisol,testosterone, growth hormone, nocturnal excretionof catecholamine and decreased sensitivity to hor-mones (Kuipers & Keizer 1988; Urhausen et al. 1995;Lehmann et al. 1997, 1999; Viru, A. & Viru 2001).Several overtraining manifestations reflect a generalimbalance in the level of hypothalamic neuro-secretion and/or in the function of the anterior pituitary gland (Keizer 1998; Lehmann et al. 1998;Hackney 1999). In accordance with the listed over-training manifestations, Fry et al. (1998) found that in weight-trained men the overtrained state isindicated by strength decrements and a pronounceddecrease in blood cortisol concentration. Overtrainedmen exhibited a slightly increased testosterone levelafter the test exercise.

In disagreement, several researchers believe that high cortisol levels in training are related todysadaptation. The reason for this opinion is thecatabolic effect of glucocorticoids that has been setin contrast to the anabolic action of testosterone.However, it should not be forgotten that in stress situations, including strenuous exercise, enhancedcatabolism is not a misfortune but ultimately a neces-sary adaptation response (creation of the pool offree amino acids to be used as additional substratefor oxidation as well as building blocks for the

adaptive protein synthesis). Moreover, during acuteexercise, cortisol makes an essential contribution to amino acid transamination, and thereby to theformation of alanine, to glyconeogenesis, to the synthesis of urea, to the synthesis of cateholaminesand supporting their effects on the post-receptorlevel, and to the control of sodium–potassium fluxesthrough cellular membrane, etc. Therefore, insuffi-cient or reverse responses of cortisol should make usthink about discrepancies in the metabolic control.

Adlercreutz et al. (1986) recommended using theratio between free testosterone and cortisol as anindication of overstrain if the ratio decreases morethan 30% or if the ratio is less than 0.35 × 10–3. Thisway, an extreme situation in the balance of anabolicand catabolic stimulation is indicated. However, theproposed quantitative measure was later frequentlyforgotten and any decrease in the ratio of testo-sterone/cortisol was considered a bad indication.With this background, conclusions on overtrainingwere made, although the authors’ data demon-strated a good performance level. Even if the ratiodecreases more than 30% or is less than 0.35 × 10–3,indications of overtraining may not exist (Kuipers &Keizer 1988; Urhausen et al. 1995).

In essence, the main shortcoming of the use of this ratio is that the hormone level in blood is not linearly related to its metabolic effects. This ratio hasbeen used despite the lack of information about thestate of the glucocorticoid and androgen receptor.Actually, the state of these receptors as well as thecompetition between testosterone and glucocortioidfor glucocorticoid receptor are essential for deter-minants of the outcome in anabolism/catabolismbalance. Consequently, the ratio of testosterone/cortisol is too indirect to be used.

Testosterone, training effects andperformance

Induction of synthesis of myofibrillar proteins

The main result of strength training is myofibrillarhypertrophy. This process is founded on the induc-tion of synthesis of myosin and actin (Booth, F.W. &Thomason 1991). While various metabolic factorsare able to induce this process, a powerful amplifier

Page 347: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 331

of the induction of synthesis of contractile proteinsis testosterone (Viru, A. 1995). In fact, amplificat-ory effect of testosterone has been evidenced by theprohibited use of anabolic steroids by athletes.Frequently, although not always, the result wasenhanced training effects in strength and powerevents (Rogozkin 1979; Wilson 1988; Lamb 1989).Rat experiments evidenced that anabolic steroidsstimulate the synthesis of myofibrillar proteins,increase RNA polymerase activity and augment the corresponding training effects (Rogozkin 1979;Rogozkin & Feldkoren 1979). Accordingly, it hasbeen assumed that in normal conditions of trainingthe synthesis of myofibrillar proteins is amplified bythe endogenous androgens.

It has been mentioned above (pp. 327–8) that,during the recovery period after exercises requiringstrong muscle contractions, a number of bindingsites increase the cytoplasm of working muscles inassociation with increased production of proteins inthe muscle (Tchaikovski et al. 1986). The signific-ance of androgen receptors in training hypertrophywas confirmed in rats: a pharmacological blockadeof androgen receptors prevented training-inducedmuscle hypertrophy (Inoue et al. 1994). At the sametime muscular activity itself increased the numberof androgen binding sites in rat muscles (Inoue et al.1993). Control of testosterone action at the receptorlevel ensures a fiber-type specific stimulation of protein synthesis in muscles during the recoveryperiod. Resistance exercises induce down-regula-tion of androgen receptors in slow-twitch fibers andup-regulation of these receptors in fast-twitch fibers(Deschenes et al. 1994). Consequently, the tissue’ssusceptibility to testosterone’s effect increases selectively during resistance exercise in fast-twitchmuscle fibers (Fig. 23.3).

In humans, the testosterone effect on musclestrength and protein synthesis has been confirmed

(Urban et al. 1995; Kraemer, W.J. et al. 1996). Whentraining induced enhanced testosterone, cortisoland growth hormone responses, the improvementof muscle strength was pronounced (Hansen et al.2001). In disagreement, Hickson et al. (1994) reportedincreased cross-sectional area of fast-twitch fibers as a result of 8 weeks of heavy resistance trainingwithout increases of testosterone basal level or its responses in exercises. However, testosteronechanges might take part during the recovery period;also possible were alterations at the receptor level.

Thyroid hormones add their action on transcrip-tion of oxidative enzymes and myosin (Freeberg &Hamolsky 1974; Konovalova et al. 1997). The actionof growth hormone, growth factors and insulin onthe translation process (Balon et al. 1990; Fryburg et al. 1991) exerts a general support for the actualiza-tion of the adaptive protein synthesis (Fig. 23.4).

Training-induced muscle hypertrophy is con-trolled at transcription, translation and post-translation level (Booth, F.W. & Thomason 1991),whereas the used exercises may determine the relative significance of contribution of actions ateach control level. Therefore, the hormonal effectson muscle hypertrophy also vary in dependence ofthe exercises used.

After exercise, the catabolic influence of cortisol is essential to provide free amino acids for proteinsynthesis, to maintain the rate of protein degrada-tion for renewal of protein structures and to adjustthe number of protein molecules to the actual need(the post-translation control) (Viru, A. 1995).

Preconditioning effect of testosterone

Data has suggested that performance in musclepower tests relates to the blood level of testosteronein athletes. In professional soccer players, the resultsof counter-movement jump positively correlated

Type I fibers

Resistance exercises

Type II fibers

Down-regulation ofandrogen receptors

Up-regulation ofandrogen receptors

Fig. 23.3 Effect of resistance exerciseson androgen receptor in muscle fiberstype I and type II.

Page 348: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

332 chapter 23

with the basal level of testosterone in blood (Bosco et al. 1996b). The specific relationship betweentestosterone level and the explosive strength of legmuscles was supported by the fact that aerobicendurance, determined by Cooper’s 12-min runstest, showed negative correlation with testosteronelevel (Bosco et al. 1996b). Comparison of testo-sterone, determined by morning samples and the riseof the center of gravity in the counter-movementtest in 97 high-level athletes, indicated the highestvalues of testosterone concentration and jumpingperformance in sprinters and the lowest values incross-country skiers, whereas soccer players exhib-ited intermediate values (Bosco & Viru 1998). Inaccordance, Kraemer, W.J. et al. (1995b) reported apositive correlation between testosterone level andperformance in double knee extension exercises.Significant correlations were also found betweenaverage power output during continuous jumping

for 60 s and the change of blood testosterone con-centration during this test (Bosco et al. 1996a).

Since the metabolic effect of testosterone is a time-consuming process due to the formation of thetestosterone-receptor complex, influence of this com-plex on the genome and synthesis of new proteins(Liao 1977; Mainwaring 1977), it is not believed that,during short-term acute force efforts, jumping exer-cise or sprint distances, necessary time is providedto activate secretion of testosterone, to transport theaugmented amount of the hormone to workingmuscle and to actualize related metabolic events.Therefore, a hypothesis has been raised that testo-sterone has a preconditioning action on the applica-tion of muscle force and power (Viru, A. & Viru2001). Competition performance (as well as the mainpart of a training sessions) is preceded by warmingup. Athletes are influenced by the anticipatory state.During competition each athlete can use three or sixtrials. Therefore, in competition power exercises areperformed on the level of altered hormone levels inblood. The significance of these hormonal alterationsfor performance depends on the time required forthe actualization of metabolic effects of hormones.The hormones, which are bound on cellular mem-brane and acting through formation of cyclic adeno-sine monophosphate (cAMP) (e.g. catecholamines)need only a couple of seconds to evoke their meta-bolic effects. The hormones, which are bound byspecific receptors in cytoplasm and acting throughthe induction of protein synthesis (e.g. testosteroneand other steroid hormones), in several cases requiremore than 1 h to show metabolic effects. Conse-quently, performance in exercises of explosive typeof power output may be influenced by hormonalchanges before the main performance.

Two types of preconditioning of the perform-ance in power events, exerted by endogenoustestosterone, may be distinguished. The long-termpreconditioning is related to the influence of tes-tosterone on the development of fast twitch (FT)muscles. Mainly, it is related to the pubertal period.The short-term preconditioning is related to theinfluence of testosterone either on the central nerv-ous system or peripheral neuromuscular apparatus.The result is ‘tuning’ of the motor centers of the cent-ral nervous system for explosive performance.

Control at the levelof transcription

Inductors

TRANSCRIPTION

TRANSLATION

InsulinGrowth hormone

Cortisol

Control at the levelof translation

Control at thepost-translationlevel

Thyroid hormonesTestosterone

Microsomes and polysomes

Genome

mRNA

New proteins

Protein degradationadjust the protein levelsto the actual necessities

Fig. 23.4 Hormonal control of synthesis of myofibrillarproteins.

Page 349: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 333

Long-term preconditioning. Explosive contractile activ-ity of muscles (jumping, sprinting performance,etc.) is related to the percentage of FT fibers in legmuscles (Costill et al. 1976; Bosco & Komi 1979).Thus, the formation of capacity for muscular activit-ies of explosive type depends on the development of FT fiber and fast motor units. Results of severalstudies indicate that testosterone is, plausibly,responsible for improved anaerobic enzyme sys-tems and structural development of FT fibers inmuscles. Bass et al. (1971) established that in tempor-alis muscle of guinea pig, sexual differentiation ofenzyme pattern might be converted by testosterone.Dux et al. (1982) demonstrated that pubertal castra-tion alters the structure of skeletal muscle. Most ofthem suffered in the development of FT muscles.Krotiewski et al. (1980) confirmed the castrationeffects in male rats. Testosterone substitutionrestored development of FT muscles in male cas-trates. According to these results, it is possible toassume that during puberty inter-individual dif-ferences in testosterone action determine the forma-tion and development of FT fibers. Several studiesin male adolescents support this assumption.Already at the onset of puberty (in 11–12-year-oldboys) area of FT fibers as well as blood lactate levelafter 15 s all-out exercise correlated significantlywith testosterone level (Mero 1988). In circum-pubertal boys testosterone levels in blood or salivacorrelated with maximal anaerobic power (Mero et al. 1990; Falgairette et al. 1991), maximal poweroutput in incremental exercise (Fahey et al. 1979),blood lactate level after Wingate test (Mero et al.1990) and maximal voluntary strength (Mero 1988).Bosco (1993) indicated that between 8.5 and 14.5years the rise of center gravity in counter movementjump increases linearly in children of both gender.From the age of 14.5 years a prevalence of boysappeared. Typical for this age is a pronouncedincrease of blood testosterone concentration.

Thus, in the pubertal period, enhanced increase of testosterone concentration in blood obviouslyfavors the development of FT fibers. Thereby a phe-notype is formed which is characterized by hightestosterone level and effective performance in exer-cise of explosive application of forces.

Besides pubertal period, inherent high levels of

testosterone may enhance myofibrillar hypertrophyin resistance training. In power events, essential isthe influence of high testosterone levels on centralnervous structures. Various influences in early post-natal life play a role in the choice of neurons of the central nervous system, which become sensitiveto steroids. The spinal nucleus of the bulboca-vernosus is highly androgen-sensitive. Testosteroneregulates both the size of motoneurons of the spinalnucleus of the bulbocavernosus and also relatedmuscle in adulthood (Kurz et al. 1986; Araki et al.1991; Lubisher & Arnold 1995). This neuromus-cular subsystem plays an important role in malecopulatory behavior. Still, there is no evidence thattestosterone influences the neural adaptations instrength training at the level of spinal motoneuronscovering the function of major muscles of the body.This may be suggested by the fact that androgensare able to influence the structure of neurons,including dendritic branching and synapse forma-tion in the adult brain (Arnold & Breedlove 1985;Matsumoto 1992). The contribution of testosteronein training-induced long-term neural adaptivityawaits investigation.

Short-term preconditioning. Another way to under-stand the significance of testosterone in power exer-cises is the short-term preconditioning effect. In theshort-term preconditioning action, the testosteronelevel in blood is, plausibly, highly significant.Investigation of aggressive behavior showed weakpositive correlations between the blood level oftestosterone and manifestations of aggressivenessin humans (Archer 1991; Book et al. 2001). Thetestosterone level is not the single determinant ofaggressive behavior, which actually depends on theinteraction of several factors, including previousexperience, environment, danger of the situation,etc. (Mazur & Booth 1998). Thus, the testosteronelevel is a preconditioning prerequisite for aggress-ive behavior but not the determinant of the expres-sion of aggressiveness. High levels of testosteronemade boys more impatient and irritable, which in turn increased their propensity to engage inaggressive–destructive behavior (Olweus et al.1988). By analogy, it is possible to assume that testo-sterone promotes changes in neurons, which are not

Page 350: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

334 chapter 23

only related to increased aggressiveness but also tofavorable mobilization of neuromuscular capacityfor explosive performance in power throwing,jumping and sprint events.

During sport competition, the short-term pre-condition effect may appear due to the increasedtestosterone concentration in blood as a result ofwarming-up exercises, anticipatory state, emotionalstrain aroused from performance of other compet-itors, as well as of prolonged concentration for theforthcoming competition. It should be rememberedthat during sports competition in judo or tennis,winners have higher levels of testosterone thanlosers (Elias 1981; Booth, A. et al. 1989). ThereforeMazur and Booth (1998) suggested that testosteroneprepared winners for effective performance.

Some anecdotic examples support the suggestionof the significance of testosterone in the precon-ditioning of performance in power events. A coupleof days before an international competition, lowtestosterone concentration was found in a discusthrower. His performance was lower than expected.Within 3 weeks following the competition his tes-tosterone concentration normalized. Further, afterreturn from winning an international competition adecathlete showed an unusually high testosteronelevel (36 mmol·L–1).

On the background of extended experimentalmaterial, Ingle (1952) affirmed that the permissiveeffect of hormones consists in enabling changes inbody function or metabolic processes (gives the per-mission to change), although the hormone itself isnot the direct cause of the change. The supposedpermissive action of testosterone is obviously re-lated to the indirect effect of testosterone, which isactualized without participation of the androgenreceptor (Nieschlag & Behre 1998). The manifesta-tions of the indirect effect of testosterone are: pro-duction of insulin-like growth factor I; competitionfor the specific binding sites of glucocorticoids;autocrine release of andromedins; transmembraneinflux of extracellular calcium; and activation ofextracellular signal-related kinase cascade via bind-ing to a yet unidentified extracellular receptor.Specification of testosterone action on muscle forceand power generation in regard of these possibilit-ies, as well as localization of related effect(s) in neu-

rons, synapses or muscle fibers is a matter of furtherinvestigation.

In conclusion. The hypothesis on the preconditioning of performance in power events by endogenoustestosterone opens a wide spectrum of tasks for fur-ther research. Perspectives include testing variousaspects of the hypothesis as well as detailed invest-igations in order to establish the cellular–metabolicfoundations for various actions of testosterone onnervous structures and muscle related to power performance.

Conclusions

Testosterone level in blood increases in men duringresistance exercises. This response depends on sev-eral conditions, among those the primary significanceseems to belong to the application of high muscleforces or power output during a sufficiently longtime (> 10–15 min) over relatively short rest pauses.During short-term resistance exercises rapid testo-sterone response is mainly related to hemoconcen-tration. After resistance training sessions a secondaryincrease of blood testosterone concentration mayappear in the late recovery period concomitantlywith increased binding of testosterone by specificsites of androgen receptors in muscle fibers. Resist-ance exercises, probably also power exercises, causethe up-regulation of androgen receptors mainly infast-twitch fibers. The most important adaptive effectof testosterone is the induction of the synthesis ofcontractile proteins, mainly in fast-twitch fibers. Thiseffect is founded on the formation of the testosterone-receptor complex and its action on the genome. Thelate recovery period after resistance training sessionsis the time for actualization of the adaptive proteinsynthesis induced by testosterone. The anaboliceffect of testosterone-initiating transcription of syn-thesis of myofibrillar proteins (release of relatedmRNA) is supported by action of growth hormoneand growth factors on the translation process.

Action of testosterone on acute muscle per-formance is also possible without the contributionof androgen receptors. The resulting preconditioneffect of testosterone is still a suggestion and re-quires systematic investigation.

Page 351: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 335

ReferencesAdlercreutz, H., Härkönen, M.,

Kuoppasalmi, K. et al. (1986) Effect of training on plasma anabolic andcatabolic steroid hormones and theirresponse during physical exercise.International Journal of Sports Medicine 7(suppl.), 27–28.

Ahtiainen, J.P., Pakarinen, A., Kraemer,W.J. & Häkkinen, K. (2003) Acutehormonal and neuromuscular responses and recovery to forced vs.maximum repetitions multipleresistance exercises. International Journal of Sports Medicine 24, 410–418.

Araki, I., Harada, Y. & Kuno, M. (1991)Target-dependent hormonal control of neuron size in the rat spinal nucleus of the bulbocavernosus. Journal ofNeurosciences 11, 3025–3033.

Archer, J. (1991) The influence oftestosterone on human aggression.British Journal of Psychology 82, 1–28.

Arnold, A.& Breedlove, M. (1985)Organizational and activational effectsof sex steroids on brain and behavior: a reanalysis. Hormones and Behavior 19,469–498.

Balon, T.W., Zorzano, A., Treadway, J.L.,Goodman, M.N. & Ruderman, N.B.(1990) Effects of insulin on proteinsynthesis and degradation in skeletalmuscle after exercise. American Journal ofPhysiology 258, E92–E97.

Bass, A., Gutmann, E., Hanzlikova, V. &Syrovy, I. (1971) Sexual differentiation of enzyme pattern and its conversion bytestosterone in temporalis muscle of theguinea pig. Physiologica Bohemoslovenica20, 423–431.

Bloomer, R.J., Sforzo, G.A. & Keller, B.A.(2000) Effects of meal form andcomposition on plasma testosterone,cortisol, and insulin following resistanceexercise. International Journal of SportNutrition, Exercise and Metabolism 10,415–424.

Book, A.S., Starzyk, K.B. & Quinsley, V.L.(2001) The relationship betweentestosterone and aggression: a meta-analysis. Aggression and Violent Behavior6, 579–599.

Booth, A., Shelley, G., Mazur, A., Thorp, G.& Kittok, R. (1989) Testosterone, and winning and losing in humancompetition. Hormones and Behavior 23,556–571.

Booth, F.W. & Thomason, D.B. (1991)Molecular and cellular adaptation ofmuscle in response to exercise:

track athletes. Journal of AppliedPhysiology 40, 149–154.

Cotshalk, L.A., Loeb, C.C., Nindl, M. et al.(1997) Hormonal responses of multisetversus singleset heavy-resistanceexercise protocols. Canadian Journal ofApplied Physiology 22, 244–255.

Cumming, D.C., Vickovic, M.M., Wall,S.R., Fluker, M.R. & Belcastro, A.N.(1985) The effect of acute exercise onpulsatile release of luteinizing hormonein women runners. American Journal ofObstetrics and Gynecology 153, 482–485.

Cumming, D.C., Wall, S.R., Galbraith,M.A. & Delcastro, A.N. (1987)Reproductive hormone responses toresistance exercise. Medicine and Sciencein Sports and Exercise 19, 234–238.

Danhaive, P.A. & Rousseau, G.G. (1988)Evidence for sex-dependent anabolicresponse to androgenic steroidsmediated by glucocorticoid receptor inrats. Journal of Steroid Biochemistry 29,275–281.

Deschenes, M.R., Maresch, C.M.,Armstrong, L.E., Kraemer, W.J. &Covault, J.M. (1994) Endurance andresistance exercise induced muscle fibertype specific responses in androgenbinding capacity. Journal of SteroidBiochemistry 50, 175–179.

Diamond, P., Brisson, G.R., Candas, B. & Perronet, F. (1989) Trait anxiety,submaximal physical exercise and bloodandrogens. European Journal of AppliedPhysiology 58, 699–704.

Di Luigi, L., Guidetti, L., Baldari, C. et al.(2002) Physical stress and qualitativegonadotropin secretion: LH biologicalactivity at rest and after exercise intrained and untrained men. InternationalJournal of Sports Medicine 23, 307–312.

Durand, R.J., Castrence, V.D., Hollander,D.B. et al. (2003) Hormonal responsesfrom concentric and eccentric musclecontraction. Medicine and Science inSports and Exercise 35, 937–944.

Dux, L., Dux, E. & Guba, F. (1982) Furtherdata on the androgenic dependence of the skeletal musculature: effect ofpubertal castration on the structure ofthe skeletal muscle. Hormonal andMetabolic Research 14, 191–194.

Elias, M. (1981) Serum cortisol,testosterone, and testosterone-bindingglobulin response to competitivefighting in human males. AggressiveBehavior 7, 215–224.

Fahey, D.T., Valle-Zuris, A.D., Oehlsen, G.,Trieb, M. & Seymour, J. (1979) Pubertal

perspectives of various models.Physiological Reviews 71, 541–585.

Bosco, C. (1993) Test di valutazione delladonna nella practica del calcio. In:Confegno Nazionale ‘Il Calcio Femminile’Aspetti Medici e Tecnici (Cambri, R. &Paterni, M., eds.). FIGC, Rome: 219–230.

Bosco, C. & Komi, P. (1979) Mechanicalcharacteristics and fiber composition of human leg extensor muscles. European Journal of Applied Physiology 41,275–284.

Bosco, C. & Viru, A. (1998) Testosteroneand cortisol levels in blood of malesprinters, soccer players and cross-country skiers. Biology of Sport 15, 3–8.

Bosco, C., Tihanyi, J., Rivalta, L. et al.(1996a) Hormonal responses instrenuous jumping effort. JapaneseJournal of Physiology 46, 93–98.

Bosco, C., Tihanyi, J. & Viru, A. (1996b)Relationship between field fitness testand basal serum testosterone andcortisol levels in soccer players. ClinicalPhysiology 16, 317–322.

Bosco, C., Colli, R., Bonomi, R., vonDuvillard, S.P. & Viru, A. (2000)Monitoring of strength training:neuromuscular and hormonal profile.Medicine and Science in Sports and Exercise32, 202–208.

Busso, T., Häkkinen, K., Pakarinen, A. et al. (1990) A systems model of trainingresponse and its relationship tohormonal responses in elite weight-lifters. European Journal of AppliedPhysiology 61, 48–54.

Busso, T., Häkkinen, K., Pakarinen, A. et al. (1992) Hormonal adaptations andmodelled responses in elite weightliftersduring 6 weeks of training. EuropeanJournal of Applied Physiology 64, 381–386.

Chwalbinska-Moneta, J., Krysztofiak, H.,Ziemba, A., Nazar, K. & Kaciuba-Uscilko, H. (1996) Threshold increase inplasma growth hormone in relation toplasma catecholamine and blood lactateconcentration in endurance-trainedathletes. European Journal of AppliedPhysiology 73, 117–120.

Copeland, J.L. Consitt, L.A., & Tremblay,M.S. (2002) Hormonal responses toendurance and resistance exercise in females 19–69 years. Journals ofGerontology. Series A, Biological Sciencesand Medical Sciences 54, B158–B165.

Costill, D.L., Daniels, J., Evans, W. et al.(1976) Skeletal muscle enzymes andfiber composition in male and female

Page 352: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

336 chapter 23

stage differences in hormonal andhematological responses to maximalexercise in males. Journal of AppliedPhysiology 46, 823–827.

Falgairette, G., Bedu, M., Fellmann, N.,VanPraagh, E. & Coudert, J. (1991)Bioenergetic profile in 144 boys aged 6 to15 years with special reference to sexualmaturation. European Journal of AppliedPhysiology 62, 151–156.

Fleck, S.J. & Kraemer, W.J. (1997) DesigningResistance Programs. Human Kinetics,Champaign, IL.

Freeberg, A.S. & Hamolsky, M.W. (1974)Effects of thyroid hormones on certainnonendocrine organ systems. In:Handbook of Physiology, Endocrinology,Thyroid. American Physiological Society,Washington, D.C.: 435–468.

Fry, A.C., Kraemer, W.J., Stone, M.H. et al. (1993) Endocrine and performanceresponses to high volume training andamino acid supplementation in elitejunior weight athletes. InternationalJournal of Sport Nutrition 3, 306–322.

Fry, A.C., Kraemer, A.C., Stone, M.H. et al. (1994) Endocrine responses tooverreaching before and after 1 year ofweightlifting. Canadian Journal of AppliedPhysiology 19, 400–410.

Fry, A.C., Kraemer, W.J. & Ramsey, L.T.(1998) Pituitary adrenal–gonadalresponses to high-intensity resistanceexercise overtraining. Journal of AppliedPhysiology 85, 2352–2359.

Fryburg, D.A., Gelfand, R.A. & Barrett, E.J. (1991) Growth hormone acutelystimulates forearm muscle proteinsynthesis in normal humans. AmericanJournal of Physiology 260, E499–E504.

Galbo, H. (1983) Hormonal and MetabolicAdaptation to Exercise. Thieme, Stuttgart,Germany.

Galbo, H., Hummel, L. Petersen, I.B.,Christensen, N.J. & Bie, W. (1977)Thyroid and testicular responses tograded and prolonged exercise in man.European Journal of Applied Physiology 36,101–106.

Galbo, H., Kjær, M. & Secher, N.H. (1987)Cardiovascular, ventilatory andcatecholmine responses to maximalexercise in partially curarized man.Journal of Physiology 389, 557–568.

Guezennec, C.Y., Legar, L., Lhoste, F.,Aymonod, M. & Pesquues, P.C. (1986)Hormones and metabolite responses to weight-lifting training sessions.International Journal of Sports Medicine 7,100–105.

Hackney, A.C. (1996) The malereproduction system and endurance

term strength training on humanskeletal muscle: the importance ofphysiologically elevated hormonelevels. Scandinavian Journal of Medicineand Science in Sports 11, 347–354.

Hickson, R.C., Hidaka, K., Foster, C.,Falduto, M.T., Chatterton, R.J. (1994)Successive time courses of strengthdevelopment and steroid hormoneresponses to heavy-resistance training.Journal of Applied Physiology 76, 663–670.

Hortobagyi, T., Houmard, J.A., Stevenson,J.R. et al. (1993) The effects of detrainingon power athletes. Medicine and Sciencein Sports and Exercise 25, 929–935.

Ingle, D.J. (1952) The role of the adrenalcortex in homeostasis. Journal ofEndocrinology 8, xxii–xxxvii.

Inoue, K., Yamasaki, S, Fushika, T., Okada,Y. & Sugimoto, E. (1993) Rapid increasein the number of androgen receptorsfollowing electrical stimulation of the rat muscle. European Journal of AppliedPhysiology 66, 134–140.

Inoue, K., Yamasaki, S., Fushiki, T., Okada,Y., Sugimoto, E. (1994) Androgenreceptor agonist suppresses exercise-induced hypotrophy of skeletal muscle.European Journal of Applied Physiology 69,88–91.

Jensen, J., Oftebro, H., Breigan, H. et al.(1991) Comparison of changes intestosterone concentration after strengthand endurance exercise in well trainedmen. European Journal of AppliedPhysiology and Occupational Physiology63, 467–471.

Jezova, D., Vigas, M., Tatár, P. et al. (1985)Plasma testosterone and catecholamineresponses to physical exercise ofdifferent intensities in men. EuropeanJournal of Applied Physiology andOccupational Physiology 54, 61–66.

Jürimäe, T.K., Karelson, K., Smirnova, T. & Viru, A. (1990) The effect of a single-circuit weight-training session on thebiochemistry of untrained universitystudents. European Journal of AppliedPhysiology and Occupational Physiology61, 344–348.

Kjær, M., Secher, N.H., Bach, F.W. &Galbo, H. (1987) Role of motor centeractivity for hormonal changes andsubstrate mobilization in humansAmerican Journal of Physiology 253,R687–R697.

Kjær, M., Secher, N.H., Bach, F.W. &Galbo, H. (1989) Hormonal andmetabolic responses to exercise inhumans: effect of sensory nervousblockade. American Journal of Physiology257, E85–E97.

training. Medicine and Science in Sportsand Exercise 28, 180–189.

Hackney, A.C. (1999) Neuroendocrinesystem. Exercise overload andregeneration. In: Overload, PerformanceIncompetency, and Regeneration in Sport(Lehmann, M., Foster, C., Gastmann, U.,Keizer, H. & Steinacker, J.M., eds.).Kluwer Academic/Plenum, New York.

Hackney, A.C., Szczepanowska, E. & Viru,A. (2003) Basal testicular testosteroneproduction in endurance-trained men issuppressed. European Journal of AppliedPhysiology 89, 198–201.

Häkkinen, K. & Pakarinen, A. (1991)Serum hormones in male strengthathletes during short term strengthtraining. European Journal of AppliedPhysiology 63, 194–199.

Häkkinen, K. & Pakarinen, A. (1993) Acutehormonal responses to two differentfatiguing heavy-resistance protocols in male athletes. Journal of AppliedPhysiology 74, 882–887.

Häkkinen, K. & Pakarinen, A. (1995) Acutehormonal responses to heavy resistanceexercise in men and women at differentages. International Journal of SportsMedicine 16, 507–513.

Häkkinen, K., Pakarinen, A., Alen, M. & Komi, P.V. (1985) Serum hormonesduring prolonged training ofneuromuscular performance. EuropeanJournal of Applied Physiology 53, 287–293.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi, P.V. (1987)Relationship between training volume,physical performance capacity, andserum hormone concentrations duringprolonged training in elite weight lifters.International Journal of Sports Medicine 8(suppl.), 61–65.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi, P.V. (1988a)Neuromuscular and hormonaladaptations in athletes to strengthtraining in 2 years. Journal of AppliedPhysiology 65, 2406–2412.

Häkkinen, K., Pakarinen, A., Alen, M.,Kauhanen, H. & Komi, P.V. (1988b)Neuromuscular and hormonalresponses in elite athletes to twosuccessive strength trainings. EuropeanJournal of Applied Physiology 57, 133–139.

Häkkinen, K., Pakarinen, A, Newton, R.U.& Kraeer, W.J. (1998) Acute hormoneresponses to heavy resistance lower andupper extremities exercise in youngversus old men. European Journal ofApplied Physiology 77, 312–319.

Hansen, S., Kvorning, T., Kjær, M. &Sjøgaard, G. (2001) The effect of short-

Page 353: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

resistance exercise and testosterone 337

Kochakian, C.D. (1935) Effect of malehormone on protein metabolism ofcastrate dogs. Proceedings of the Society for Experimental Biology and Medicine 32,1064–1065.

Kochakian, C.D. (1959) Mechanisms of androgen action. LaboratoryInvestigations 8, 538–556.

Kochakian, C.D., Costa, G. & Hill, J. (1964)Effect of testosterone propionate onmetabolism and incorporation ofglysine-2-C 14 in guinea pig tissuepreparations. Acta Endocrinologica 46(suppl.), 92.

Konovalova, G., Masso, R., Ööpik, V. &Viru, A. (1997) Significance of thyroidhormones in post-exercise incorporationof amino acids into muscle fibers: anautoradiographic study. Endocrinologyand Metabolism 4, 25–31.

Krabbe, S., Transbøl, I. & Cristensen, C.(1982) Bone mineral homeostasis, bonegrowth and mineralization during yearsof pubertal growth: a unifying concept.Archives for Diseases of Childhood 57,359–375.

Kraemer, R.R., Heleniak, R.J., Tryniecki,J.L. et al. (1995) Follicular and lutealphase hormonal responses to low-volume resistance exercise. Medicine andScience in Sports and Exercise 27, 809–817.

Kraemer, W.J., Patton, J.F., Knuttgen, H.G.et al. (1989) Hypothalamic–pituitary–adrenal responses to short-durationhigh-intensity cycle exercise. Journal ofApplied Physiology 66, 161–166.

Kraemer, W.J., Marchitelli, L., Gordon, S.E.et al. (1990) Hormonal and growth factorresponses to heavy resistance protocols.Journal of Applied Physiology 69, 1442–1250.

Kraemer, W.J., Gordon, S.E., Fleck, S.J. et al.(1991a) Endogenous anabolic hormonaland growth factor responses to heavyresistance exercise in males and females.International Journal of Sports Medicine 21,228–235.

Kraemer, W.J., Patton, J.F., Knuttgen, H.G.et al. (1991b) Effects of high-intensitycycle exercise on sympathoadrenal–medullary response pattern. Journal ofApplied Physiology 70, 8–14.

Kraemer, W.J., Fry, A.S., Warren, B.J. et al.(1992) Acute hormonal responses in elitejunior weightlifters. International Journalof Sports Medicine 13, 103–109.

Kraemer, W.J., Fleck, S.J., Dziados, J.E. et al. (1993) Changes in hormoneconcentrations after different heavy-resistance exercise protocol in women.Journal of Applied Physiology 75, 594–604.

Kraemer, W.J., Patton, J.F., Gordon, S.E. et al. (1995a) Compatibility of high-

Lamb, R.D. (1989) Anabolic steroids andathletic performance. In: Hormones andSport (Lamb, R.D., ed.). Raven Press,New York: 257–273.

Lehmann, M., Keul, L., Huber, G. & Prada,M. (1981) Plasma catecholamines intrained and untrained volunteers duringgraduated exercise. International Journalof Sports Medicine 2, 143–147.

Lehmann, M., Foster, J.M., Steinacker, J.M.et al. (1997) Training and overtraining:overview and experimental results.Journal of Sports Medicine and PhysicalFitness 37, 7–17.

Lehmann, M., Foster, C., Dickhuth, H. & Gastmann, U. (1998) Autonomicimbalance hypotheses and overtrainingsyndrome. Medicine and Science in Sportsand Exercise 30, 1140–1145.

Lehmann, M., Foster, C., Gastmann, U.,Keizer, H. & Steinacker, J.M. (1999)Definition, types, symptoms, findings,underlying mechanisms, and frequencyof overtraining and overtrainingsyndrome. In: Overload, PerformanceIncompetency, and Regeneration in Sport(Lehmann, M., Foster, C., Gastmann, U.,Keizer, H. & Steinacker, J.M., eds.).Kluwer Academic/Plenum, New York:1–6.

Liao, S. (1977) Molecular action ofandrogens. In: Biochemical Action ofHormones (Litwack, G., ed.). AcademicPress, New York: 351–406.

Lubisher, J.L. & Arnold, A.P. (1995)Evidence for target regulation of thedevelopment of androgen sensitivity inrat spinal motoneurons. Development ofNeurosciences 17, 106–117.

McCall, G.E., Byrnes, W.C., Fleck, S.J.,Dickinson, A. & Kraemer, W.J. (1999)Acute and chronic hormonal responsesto resistance training designed topromote muscle hypertrophy. CanadianJournal of Applied Physiology 24, 96–107.

Mainwaring, W.I. (1977) The Mechanism ofAction of Androgens. Spinger-Verlag,New York.

Matsumoto, A. (1992) Sex steroidinduction of synaptic reorganization inthe adult neuroendocrine brain. Reviewsof Neurosciences 3, 287–306.

Mayer, M. & Rosen, F. (1977) Interaction ofglucocortioids and androgens withskeletal muscle. Metabolism 26, 937–962.

Mazur, A. & Booth, A. (1998) Testosteroneand dominance in human males.Behavior and Brain Sciences 21, 353–397.

Mero, A. (1988) Blood lactate productionand recovery from anaerobic exercise intrained and untrained boys. EuropeanJournal of Applied Physiology 57, 660–666.

intensity strength and endurancetraining on hormonal and skeletalmuscle adaptations. Journal of AppliedPhysiology 78, 976–989.

Kraemer, W.J., Häkkinen, K, Newton, R.U.et al. (1995b) Factors in various strengthand power performance in man. In:Proceedings of the XVth Congress of theInternational Society of Biomechanics.University of Jyväskylä, Jyväskylä,Finland: 508–509.

Kraemer, W.J., Fleck, S.J. & Evans, W.J.(1996) Strength and power training:physiological mechanisms ofadaptation. Exercise and Sports ScienceReviews 24, 363–397.

Kraemer, W.J., Häkkinen, K., Newton, R.U.et al. (1998a) Acute hormonal responsesto heavy resistance exercise in youngerand older men. European Journal ofApplied Physiology 77, 206–211.

Kraemer, W.J., Volek, J.C., Bush, J.A.,Putukian, M. & Sebastianelli, W.J.(1998b) Hormonal responses toconsecutive days of heavy-resistanceexercise with or without nutritionalsupplementum. Journal of AppliedPhysiology 85, 1544–1555.

Kraemer, W.J., Konziris, L.P., Ratamess,N.A. et al. (2002) Detraining producesminimal changes in physicalperformance and hormonal variables inrecreationally strength-trained men.Journal of Strength Conditioning Research16, 373–382.

Krotiewski, M., Kral, J.G. & Karlsson, J.(1980) Effects of castration andtestosterone substitution on bodycomposition and muscle metabolism inrats. Acta Physiologica Scandinavica 109,233–237.

Kuipers, H., & Keizer, H.A. (1988)Overtraining in elite athletes: reviewand directions for the future. SportsMedicine 6, 79–92.

Kuoppasalmi, K. (1980) Plasmatestosterone and sex hormone bindingglobulin in physical exercise.Scandinavian Journal of Clinical andLaboratory Investigations 40, 411–418.

Kuoppasalmi, K., Näveri, H., Härkönen,M. & Adlercreutz, H. (1980) Plasmacortisol, androstendione, testosterone,and lutenizing hormones in runningexercise of different intensities.Scandinavian Journal of Clinical andLaboratory Investigations 40, 403–409.

Kurtz, E.M., Sengelaub, D.R. & Arnold,A.P. (1986) Androgens regulate thedendritic length of mammalianmotoneurons in adulthood. Science 232,395–398.

Page 354: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

338 chapter 23

Mero, A, Jaakola, L. & Komi, P.V. (1990)Serum hormones and physicalperformance capacity in young boyathletes during 1-year training period.European Journal of Applied Physiology 60,32–37.

Nieschlag, E. & Behre, H.M. (1998)Testosterone Action, Deficiency, andSubstitution. Springer-Verlag, New York.

Nindl, B.C., Kraemer, W.J., Gotschalk, L.A.et al. (2001) Testosterone response afterresistance exercise in women: influenceof regional fat distribution. InternationalJournal of Sports Nutrition and ExerciseMetabolism 11, 451–465.

Olweus, D., Mattson, Å., Schalling, D. &Löw, H. (1988) Circulating testosteronelevels and aggression in adolescentmales: a causal analysis. PsychosomaticMedicine 50, 261–272.

Op’Teijnde, B. & Hespel, P. (2001) Short-term creatine supplementation does notalter the hormonal response to resistancetraining. Medicine and Science in Sportsand Exercise 33, 449–453.

Padron, R.S., Wischussen, J. & Hudren, B.(1980) Prolonged biphasic response of plasma testosterone to singleintramuscular injection of humanchoronic gonadotropin. Journal ofClinical Endocrinology 50, 1100–1104.

Palacios, A., Campfield, L.A., McClure,R.D., Stein, B. & Swerdoloff, R.S. (1983)Effects of testosterone enanthate onhematopoiesis in normal men. Fertilityand Sterility 40, 100–104.

Papanicolaou, G.N. & Falk, E.A. (1938)General muscular hypertrophy inducedby androgenic hormones. Science 87,238–239.

Port, K. (1991) Serum and saliva cortisol responses and blood lactateaccumulation during incrementalexercise testing. International Journal ofSport Medicine 12, 490–494.

Potteinger, J.A., Judge, L.W., Cerny, J.A. &Pottinger, V.M. (1995) Effects of alteringtraining volume and intensity on bodymass, performance, and hormonalconcentrations in weight-event athletes.Journal of Strength Conditioning Research9, 55–58.

Pullinen, T., Mero, A., MacDonald, E.,Pakarinen, A. & Komi, P.V. (1998)Plasma catecholmine and serumtestosterone responses to four units ofresistance exercise in young and adultmale athletes. European Journal of AppliedPhysiology 77, 413–420.

Rahkila, P., Hakala, E., Alén, M., Salminen, K. & Laatikainen, T. (1988)Beta-endorphin and corticotropin

(1992) Stability and variability inhormone responses to prolongedexercise. International Journal of SportsMedicine 13, 230–235.

Viru, A., Smirnova, T., Karelson, K.,Snegovskaya, S. & Viru, M. (1996)Determinants and modulators ofhormonal responses in exercise. Biologyof Sport 13, 169–187.

Viru, A., Laaneots, L., Karelson, K.,Smirnova, T. & Viru, M. (1998) Exercise-induced hormone responses in girls atdifferent stages of sexual maturation.European Journal of Applied Physiology 76,801–805.

Viru, M. & Viru, A. (2000) HormonelleVeränderungen in der Taper. Phaseumittelbar vor einem Wettkampf.Leisungssport 30(5), 4–7.

Volek, J.S., Boetes, M., Bush, J.A. et al.(1997a) Response of testosterone andcortisol concentration to high intensityresistance exercise following creatinesupplementation. Journal of StrengthConditioning Research 11, 182–187.

Volek, J.S., Kraemer, W.J., Bush, J.A.,Incedon, T. & Boete, M. (1997b)Testosterone and cortisol in relation todietary nutrients and resistance exercise.Journal of Applied Physiology 82, 49–54.

Weiss, L.W., Cureton, K.J. & Thomson,F.N. (1983) Comparison of serumtestosterone and androstendioneresponses to weight lifting in men andwomen. European Journal of AppliedPhysiology 50, 413–419.

Weltman, J.A., Seip, R.L., Weltman, A.et al. (1990) Release of lutenizinghormone and growth hormone afterrecovery from maximal exercise. Journalof Applied Physiology 69, 196–200.

Wierman, M.E. & Crowley, W.F. (1986)Neuroendocrine control of the onset of puberty. In: Human Growth: AComprehensive Treatise, vol. 2. PostnatalGrowth: Neurobiology, 2nd edn. (Falkner,F. & Tanner, J.M., eds.). Plenum, NewYork: 225–241.

Wilkerson, J.E., Horwath, S.M. & Gutin, G.(1980) Plasma testosterone duringtreadmill exercise. Journal of AppliedPhysiology 49, 249–253.

Wilson, J.D. (1988) Androgen abuse byathletes. Endocrinological Review 9,181–199.

Youl, K.H., Hwak, K.S., JunLee, W. &Bryne, H.K. (2002) Effects of ginsengingestion on growth hormone,testosterone, cortisol, and insulin-likegrowth factor 1 responses to acuteresistance exercise. Journal of StrengthConditioning Research 16, 179–183.

release is dependent on a thresholdintensity of running exercise in maleendurance athletes. Life Sciences 43,551–558.

Ritzen, M., Aperia, A., Hall, K. et al. (eds)(1981) The Biology of Normal HumanGrowth. Raven Press, New York.

Rogozkin, V.A. (1979) Metabolic effects ofanabolic steroids. Medicine and Science inSports 11, 160–163.

Rogozkin, V.A. & Feldkoren, B.I. (1979)The effect of retabolil and training onactivity of RNA polymerase in skeletalmuscle. Medicine and Science in Sports 11,345–347.

Schwab, R., Jounson, G.O., Housh, T.J.,Kinder, J.E. & Weir, J.P. (1993) Acuteeffects of different intensities of weightlifting on serum testosterone. Medicineand Science in Sports and Exercise 25,1381–1385.

Scow, R.V. & Hagen, S.H. (1955) Effect of testosterone propionate on myosin,collagen and other protein fractions instriated muscle of gonadectomized male guinea pigs. American Journal ofPhysiology 180, 31–36.

Sejersted, O.M., Vøllenstad, N.K. &Medbø, J.I. (1986) Muscle fluid andelectrolyte balance during and followingexercise. Acta Physiologica Scandinavica128 (suppl. 556), 119–127.

Sutton, J.R., Coleman, M.J. & Casey, J.H.(1978) Testosterone production rateduring exercise. In: Third InternationalSymposium on Biochemistry of Exercise(Landry, F. & Orban, W.A., eds.).Symposia Specialists, Miami: 227–234.

Tchaikovski, V.S., Astratenkova, I.V. &Bashirina, O.B. (1986) The effect ofexercise on the content of receptor of thesteroid hormone in rat skeletal muscle.Journal of Steroid Biochemistry 24,251–253.

Urban, R.J., Bodenburg, Y.H., Gilkson, C.et al. (1995) Testosterone administrationto elderly man increases skeletal musclestrength and protein synthesis. AmericanJournal of Physiology 269, E820–E826.

Urhausen, A., Gabriel, H. & Kindermann,W. (1995) Blood hormones as markers oftraining stress and overtraining. SportsMedicine 30, 351–376.

Viru, A. (1992) Plasma hormones andphysical exercise. International Journal ofSports Medicine 13, 201–209.

Viru, A. (1995) Adaptation in SportsTraining. CRC Press, Boca Raton, FL.

Viru, A. & Viru, M. (2001) BiochemicalMonitoring of Sport Training. HumanKinetics, Champaign, IL.

Viru, A., Karelson, K. & Smirnova, K.

Page 355: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

339

Introduction

β-Endorphin (βE) and cortisol are two importantneurohormones that influence immune responseand glucose levels that ultimately arise from a com-mon molecule. This common molecule (preprohor-mone) proopiomelanocortin (POMC) (Fig. 24.1) canbe cleaved to several peptide components. POMC is not only the precursor of adrenocorticotrophichormone (ACTH) that stimulates the production ofcortisol in the adrenals but also contains the peptideβE. The production of POMC and ultimately bothcortisol and βE are regulated by factors that arisefrom the hypothalamus and the paraventricularnucleus (PVN) in the brain. Corticotropin-releasinghormone (CRH) arises from the hypothalamus andis the major stimulant to activate the release ofACTH from the anterior pituitary gland. Argeninevasopression, which arises from the PVN, is also anactivator of ACTH release into the circulation. Thereare numerous factors that can alter the release ofPOMC such as diurnal, emotional, physical and bio-chemical signals. Circulating cortisol will feedbackand inhibit the production of POMC although other

molecules can influence its synthesis. βE is also synthesized within the brain and spinal cord, haspotent opioid actions within the central nervoussystem and appears to modulate pain.

βE released into the circulation arises primarilyfrom the anterior pituitary gland. The large peptidePOMC has a section towards the C-terminus knownas β-lipotropin that ultimately is cleaved to γ-lipotropin and βE. Both β- and γ-lipotropin moleculeshelp to mobilize lipid molecules from adipose tis-sue. βE within the circulation has been implicated ina number of processes including modulation ofimmune function, pain modulation and assisting inglucose homeostasis. βE receptors have been identi-fied in numerous sites in the body including adiposetissue, pancreas and skeletal muscle. However, theexact role(s) βE may have on these tissues is stillbeing elucidated.

Cortisol, the major glucocorticoid, has a negativefeedback impact on its own secretion at the level of the anterior pituitary and the hypothalamus.Cortisol acts by binding to a cytosolic receptor, andthis complex is then moved to the nucleus where itbinds to modulate gene expression. In this manner,

Chapter 24

Exercise Response of β-Endorphin and Cortisol:Implications on Immune Function

ALLAN H. GOLDFARB

Anteriorlobe ofpituitary

C

N-terminal peptide (1–76) J-peptide(1–30)

ACTH (1–39) γ -LPH 1–56 βE 58–89

N-terminal peptideN J-peptide

POMCSignal peptide

ACTH (1–39) β-LPH (1–89)

Fig. 24.1 Proopiomelanocortin (POMC) in the pituitary. Anterior pituitary lobe of pituitary. ACTH, adrenocorticotrophichormone; βE, β-endorphin; β-LPH, β-lipotropin; γ-LPH, γ-lipotropin; J, joining peptide. The numbers coincide to theamino acid sequence for that section.

Page 356: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

340 chapter 24

cortisol is the major inhibitor of the synthesis ofCRH and the synthesis of POMC. Additionally, cortisol has been shown to inhibit the release of syn-thesized ACTH stored in vesicles within the anter-ior pituitary gland. There is a control of cortisol atthe level of the hypothalamus. CRH control fromthe hypothalamus demonstrates a circadian andpulsatile nature that manifests a pulsatile and variedresponse throughout the day for these factors. The greatest pulsatile secretion of ACTH typicallyoccurs in the early morning. It should be noted thatthe suprachiasmatic nucleus of the hypothalamusreceives input from the optic nerve and this inputhas an impact on this circadian rhythm. Eliminationof the optic nerve can eliminate the circadianrhythm of ACTH and cortisol.

The formation of cortisol from cholesterol withinthe adrenal cortex takes some time. Therefore, therewill be a delay in the cortisol peak compared to theincreased pulsatile peaks of ACTH. The primaryfunction of cortisol is to help maintain glucose levelsby enhancing mobilization of amino acids from pro-teins to the liver to be precursors for glucose. Thestimulation of gluconeogenesis by cortisol as well asthe stimulation of fat mobilization to enhance fatmetabolism helps to raise plasma glucose levels.Cortisol also acts as an immunosuppressive agentand has anti-inflammatory activity.

Exercise influence on circulating ββ-endorphin and cortisol

βE increases within the circulation have been docu-mented in response to various aerobic and anaero-bic exercises (Goldfarb & Jamurtas 1997). Severalstudies have reported that circulating βE immuno-reactivity can increase in response to exercisedepending on the intensity of exercise (McMurray et al. 1987; Goldfarb et al. 1990; Kraemer, W.J. et al.1993). It appears that a critical minimum intensity of> 60% of Vo2max is needed to result in βE elevationwith aerobic exercise (McMurray et al. 1987; Goldfarbet al. 1990, 1991; Rahkila & Laatikainen 1992). How-ever, this minimum may differ based on the indi-vidual (Viru & Tendzegolskis 1995; Heitkamp et al.1996) and nutritional status of the subject. Addition-ally, the duration of the exercise appears to influ-

ence the βE response to exercise (Goldfarb et al.1990; Heitkamp et al. 1996).

Incremental exercise and high intensity anaerobicexercise has been reported to stimulate βE increaseswithin the circulation (Metzger & Stein 1984; Farrellet al. 1987; Goldfarb et al. 1987; Heitkamp et al. 1996).Resistance exercise as a stimulus to circulating βE islimited. Conflicting reports exist and may be relatedto differences in subjects, type of exercise intensityand time of measurement. Kraemer, W.J. et al. (1993)reported that βE levels in the circulation increasedin response to high total workload. These authorsnoted that the total work, rest to work ratio and totalforce needed probably influenced the βE response.These authors also reported an increase in βE in 28elite male weightlifters after a moderate to highintensity workload (Kraemer, W.J. et al. 1992). βElevels were reported to be elevated in females inresponse to three sets of resistance at 85% of their 1-repetition maximum (1-RM) (Walberg-Rankin et al.1992). An increased βE/β-lipotrophin level wasreported in response to weightlifting in five men(Elliot et al. 1984). In contrast, low volume resistanceexercise did not result in any change in βE levels(Kraemer, R.R. et al. 1996). It appears that resistanceexercise of sufficient intensity and volume (work-load) can result in a transient increase in βE levelswithin the circulation.

Cortisol response to exercise has varied depend-ing on the type of exercise, the intensity of exerciseand the duration of the exercise. Generally, mildintensity moderate duration aerobic exercise doesnot appear to alter the cortisol level within the cir-culation, although some reports have indicated adecline in cortisol. In contrast, longer duration exer-cise and more intense exercise have typically elev-ated circulating cortisol levels. This may be relatedin part, to glucose homeostasis. When individualsare given carbohydrate during long duration exer-cise the cortisol response is attenuated. Exercise of sufficient duration generally will demonstrate an increase in circulating cortisol (Galbo 1983;Petraglia et al. 1988). Cortisol levels increased in athletes who ran 1500 m and 10 000 m but not in athletes whom competed in sprints (100 m) or per-formed the discus throw (Petraglia et al. 1988).Short-term exercise may have only minor changes

Page 357: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise β-endorphin and cortisol immune function 341

in the cortisol level in plasma (Galbo 1983). It shouldbe noted that because of diurnal variations in cor-tisol levels, the increases are not always observed.

Cortisol response to resistance exercise has equiv-ocal responses partly due to the intensity of the exer-cise and also related to total workload. In a recentstudy, it was reported that force repetition exerciseincreased plasma cortisol levels to a greater extentthat maximal isotonic exercise (Ahtiainen et al.2003). The results suggest that the workload has an influence on the cortisol response. In support of this finding, resistance exercise using differentintensities of 1-RM and varying sets was shown toinfluence the cortisol response (Smilios et al. 2003).The results suggested that cortisol response wassimilar in the high intensity exercise independent ofnumber of sets whereas in lower intensity resistanceexercise four sets increased cortisol more so thantwo sets. High total work-resistance exercise wasreported to increase cortisol as well as βE (KraemerW.J. et al. 1993). It is interesting to note that the ele-vation in cortisol was fairly rapid and occurred bythe middle of the exercise as well as immediatelyfollowing the activity and during the 15-min recov-ery period. Not all studies confirm that cortisol levels will increase with high intensity exercise(Volek et al. 1997). These equivocal findings in corti-sol in response to resistance exercise may be relatedin part to diurnal variations, nutritional factors andtraining status of the subjects.

ββ-Endorphin and immune function

βE influence on immune function has been investig-ated in vitro but has not been adequately investig-ated in vivo. βE (both rat and human) was shown tostimulate T-lymphocyte proliferation (Hemmick &Bidlack 1990). The data suggests that βE mode ofaction was not through an opioid receptor but mightbe through the inhibition of the prostaglandin E1effect on immune function. Synthetic βE was shownto bind to non-opioid receptors on T-lymphocytesand was not blocked by naloxone or Met-enkephalin(Navolotskaya et al. 2001).

βE was shown in vitro to stimulate rat spleen lym-phocytes by enhancing the proliferative response toseveral mitogens (Gilman et al. 1982). The response

was dose dependent and was not blocked by naloxone. Additionally, there was no βE effect on B-lymphocytes. The proliferative response of spleno-cytes of adult male F344 rats to βE was enhanced50–100% in a dose-dependent manner on T-cells(van den Bergh et al. 1991). It was noted that inter-leukin 2 (IL-2) was elevated as well as IL-2 receptorexpression prior to the βE proliferative effect on theT-cells. Additionally, naloxone was not effective inblocking the βE effect. Further evidence on the βEproliferative effect on human T-lymphocytes wasshown using the mitogen concanavalin A (Owen et al. 1998). βE stimulated the mitogen response by121–750% with a bell-shaped curve indicating thattoo high a dose would actually inhibit the response.It also appears that this response may change withtime, dose or mitogen used (Millar et al. 1990). Theseauthors also noted that the inhibition of the immuneresponse to cortisol maybe partially reversed by βE.Therefore, the activation of βE may inhibit suppres-sion of the immune response by cortisol in vivo.

The effect of βE on human natural killer cell func-tion was studied in vitro and was enhanced when βE was present (Kay et al. 1984). They also reportedthat this was a dose dependent response and wasinhibited by naloxone. This suggests that the modeof action on natural killer cells appears to be differ-ent than the enhancement of T-lymphocyte func-tion. The βE effect on natural killer cell activity(NKCA) and amount with exercise was also studied(Gannon et al. 1998). Naltrexone (an opioid-blockingagent) was given 60 min prior to a 2-h moderateintensity exercise (65% Vo2max) and compared to aplacebo trial. The βE levels in the blood increased at90 and 120 min with exercise and NKCA and countswere elevated. Furthermore, the naltrexone treat-ment did not alter the exercise response in NKCA orcounts. These authors suggested that βE probablywas not involved in the NKCA increase with exer-cise. However, it is possible that βE may work inde-pendent of this receptor action ( Jonsdottir et al.2000). Chronic exercise (wheel running for 5 weeks)in spontaneously hypertensive rats enhanced NKCA.The βE levels in cerebrospinal fluid increased afterthe running and also enhanced clearance of lym-phoma cells from the lungs. The δ-receptor antag-onist naltrindole significantly but not completely

Page 358: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

342 chapter 24

inhibited the enhanced NKCA after 5 weeks of exer-cise. Neither κ- nor µ-receptor antagonists influ-enced the natural killer cell response. These authorssuggested a central δ-receptor mediated response to the exercise training occurred. Peripheral βEgiven subcutaneously did not alter NKCA in vivo( Jonsdottir et al. 1996). In contrast, NKCA after central injection of a δ-opioid receptor agonist wasenhanced (Band et al. 1992). In addition, a singleinjection into the intracerebral ventricle of a µ-agonist reduced NKCA. Furthermore, a single morphine injection into the periaquaduct area sup-pressed NKCA (Weber & Pert 1989). This suggeststhat central mediated βE may act to modulateNKCA via both µ-receptors and δ-receptors. Clearlymore research with training programs is needed to substantiate the training response in other populations.

Additional modes of action of βE on the immuneresponse include mononuclear cell chemotaxis (vanEpps & Saland 1984; Pasnik et al. 1999), immuno-globulin migration (van Epps & Saland 1984; Salandet al. 1988) and lymphokine production (van Epps & Saland 1984). Macrophages showed migration toβE injected into the cerebral ventricles in rats (vanEpps & Saland 1984). Human neutrophils demon-strated enhanced migration to βE infusion and theresponse was blocked by prior incubation withnaloxone. Analogs of opioids appear to have dif-ferent responses when injected into the cerebralventricles (Saland et al. 1988). Some may stimulatemacrophages and others may influence neutrophils.The chemotaxis response appears to be dose de-pendent (Pasnik et al. 1999). High doses of βE (10–3 mol) inhibited the chemotaxis response whereaslower concentrations stimulated up-regulation ofneutrophils. Since, physiological βE concentration is below the high dose level even when elevated by exercise or other stressors, it is probable βEwould have a stimulatory influence on this immunefunction.

It has been suggested that the opioid peptidessuch as βE and the enkephalins have a similar struc-tural component of interleukin-2 (Jiang et al. 2000).Interleukin-2 and other interleukins are involved inthe inflammatory response and are targets of βE andcortisol. It is highly likely that both βE and cortisol

influence the immune response by interacting withinterleukins (Zitnik et al. 1994). The inhibition maybe at a number of levels including the reduction of production of interleukin-1 and 6 in a dose-dependent manner. βE stimulated the production of interferon-γ (IFN-γ) from human mononuclearcells in response to concanavalin A in cultured cells (Brown & van Epps 1986). IFN-γ was enhancedin a dose-dependent manner using physiological βE(10–14–10–10 mol) and was not blocked by naloxone.

It appears that βE may act on a number ofimmune factors both centrally and in the peripheryand may act through both opioid and non-opioidreceptors. Additionally, the action of βE may workthrough direct interaction with cortisol.

Cortisol and immune function

Cortisol has been generally thought of as animmunosupressent and anti-inflammatory agent.Corticosteroids given intravenously to humans can induced lymphocytopenia, monocytopenia andneutrophilia but may take up to 4 h to peak (Rabin et al. 1996). High doses of corticosteroids can resultin cell death of immature T- and B-lymphocytes(Cohen & Duke 1984). Cortisol can regulate theimmune system by induction of apoptosis of thymus and blood lymphocytes (Hirano et al. 2001).However, the metabolite of cortisol oxidation, cor-tisone can inhibit the apoptosis of these lympho-cytes. Human monocyte cell death was shown to beenhanced by glucocorticoids (Schmidt et al. 1999).There was both a time and dose-dependent effect onmonocyte apoptosis. IL-1 mediated activation waspartially responsible for the enhanced monocyteapoptosis. In addition, cortisol can inhibit tumornecrosis factor-α (TNF-α) and prostaglandin E2(PGE2) by activated monocytes/macrophages (Hartet al. 1990). Interleukin-1 may also feedback to thehypothalamus to stimulate ACTH and cortisol syn-thesis (Besedovsky & del Rey 1987). Incubation of thymocytes and splenocytes with corticosteronein vitro resulted in apoptosis and necrosis of thesecells after 24 h (Hoffman-Goetz & Zajchowski 1999).The concentration of corticosterone in the mediumwas similar to the level that near maximal exer-cise would produce. This suggests that cortisol may

Page 359: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise β-endorphin and cortisol immune function 343

contribute to the apoptosis of lymphocytes and re-duced immunity after the intense exercise.

Corticosteroids have been shown to inhibitNKCA in vitro (Parillo & Fauchi 1978; Pedersen et al.1984). NKCA was shown to decrease in blood lym-phocytes after intravenous methylprednisolonepulse therapy in eight rheumatoid arthritic patients(Pedersen et al. 1984). Both in vitro and in vivo NKCAdecreased in response to cortiocosteroids (Parillo & Fauchi 1978). However, the response in vivo wasdifferent compared to in vitro. Adhesion of naturalkiller cell function to target cells was inhibited inresponse to pharmacological doses of corticosteroidsin vitro (Hoffman et al. 1981; Pedersen & Beyer 1986).The natural killer cell activity of mononuclear cellswas inhibited by methylprednisolone and hydro-cortisone in a dose-dependent manner and inhib-ited adhesion to target cells (Pedersen & Beyer1986). Inhibition of adhesion to target cells by corti-costeroids in mononuclear cells was dose depend-ent and was related to alteration in the methylationof phospholipids (Hoffman et al. 1981).

The decline in immune function associated with

cortisol will have a time delay of several hours. Theinfluence of cortisol in response to moderate or lowintensity exercise appears to be minimal on immunefunction. However, intense exercise can induce thecortisol response and this has an impact on immunefunction. In addition, chronic high intensity exercisecan reduce immune function (Pedersen & Hoffman-Goetz 2000).

Summary

Both βE and cortisol influence the immune function,with βE generally enhancing immune function andcortisol acting as an immunosupressent. The inter-play of βE and cortisol in regulating immune func-tion in response to both acute and chronic exercisestill needs clarification. The exercise effect on centralmediated βE and in vivo immune function also needsclarification. Adaptation effects to training alsoneed further study. In addition, nutritional factors(i.e. carbohydrate level and antioxidants) have notbeen adequately examined in relation to both βEand cortisol influence on the immune response.

ReferencesAhtiainen, J.P., Pakarinen, A., Kraemer,

W.J. & Häkkinen, K. (2003) Acutehormonal and neuromuscular responsesand recovery to forced vs. maximumrepetitions multiple resistance exercises.International Journal of Sports Medicine24(6), 410–418.

Band, L.C., Pert, A., Williams, W. et al.(1992) Central µ-opioid receptorsmediate suppression of natural killer activity in vivo. Progress inNeuroendocrinimmunology 5, 95–101.

van den Bergh, P., Rozing, J. &Nagelkerken, L. (1991) Two opposingmodes of action of β-endorphin onlymphocyte function. Immunology 72(4),537–543.

Besedovsky, H. & del Rey, A. (1987)Neuroendocrine and metabolicresponses induced by interleukin-1.Journal of Neuroscience Research 18(1),172–178.

Brown, S.L. & van Epps, D.E. (1986) Opioid peptides modulate production of interferon γ by human mononuclearcells. Cell Immunology 103(1), 19–26.

Cohen, J.J. & Duke, R.C. (1984)Glucocorticoid activation of a

calcium-dependent endonulease inthymocyte nuclei leads to cell death.Immunopharmacology 132, 38–42.

Elliot, D.L., Goldberg, L., Watts, W.J. &Orwoll, E. (1984) Resistance exercise and plasma β-endorphin/β-lipotrophinimmunoreactivity. Life Sciences 34(6),515–518.

van Epps, D.E. & Saland, L. (1984) Beta-endorphin and met-enkephalinstimulate human peripheral bloodmononuclear cell chemotaxis. Journal ofImmunology (Baltimore, Md.) 132(6),3046–3053.

Farrell, P.A., Kjaer, M., Bach, F.W. & Galbo, H. (1987) Beta-endorphin andadrenocorticotropin response tosupramaximal treadmill exercise intrained and untrained males. ActaPhysiologica Scandinavica 30(4), 619–625.

Galbo, H. (1983) Hormonal and MetabolicAdaptation to Exercise. Thieme Verlag,New York.

Gannon, G.A., Rhind, S.G., Suzui, M. et al.(1998) Beta-endorphin and natural killercell cytolytic activity during prolongedexercise. Is there a connection? American

Journal of Physiology 275(6 Pt 2),R1725–R1734.

Gilman, S.C., Schwartz, J.M., Milner, R.J.,Bloom, F.E. & Feldman, J.D. (1982) Beta-endorphin enhances lymphocyteproliferative responses. Proceedings of theNational Academy of Sciences of the UnitedStates of America 79(13), 4226–4230.

Goldfarb, A.H. & Jamurtas, A.Z. (1997)Beta-endorphin response to exercise: An update. Sports Medicine 24(1), 8–16.

Goldfarb, A.H., Hatfield, B.D., Sforzo, G.A.& Flynn, M.G. (1987) Serum β-endorphin levels during a gradedexercise test to exhaustion. Medicine andScience in Sports and Exercise 19(2), 78–83.

Goldfarb, A.H., Hatfield, B.D., Armstrong,D. & Potts, J. (1990) Plasma β-endorphinconcentration: response to intensity andduration of exercise. Medicine and Sciencein Sports and Exercise 22, 241–244.

Goldfarb, A.H., Hatfield, B.D., Potts, J. &Armstrong, D. (1991) Beta-endorphintime course response to intensity ofexercise: effect of training status.International Journal of Sports Medicine12(3), 264–268.

Hart, P.H., Whitty, G.A., Burgess, D.R.,

Page 360: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

344 chapter 24

Croatto, M. & Hamilton, J.A. (1990)Augmentation of glucocorticoid actionon human monocytes by interleukin-4.Lymphokine Research 9(2), 147–153.

Heitkamp, H-Ch., Schmid, K. & Scheib, K. (1996) Beta-endorphin andadrenocorticotrophic hormoneproduction during marathon andincremental exercise. European Journal ofApplied Physiology 66(3), 269–274.

Hemmick, L.M. & Bidlack, J.M. (1990)Beta-endorphin stimulates rat Tlymphocyte proliferation. Journal ofNeuroimmunology 29(1–3), 239–248.

Hirano, T., Horigome, A., Takatani, M. &Oka, K. (2001) Cortisone counteractsapoptosis-inducing effect of cortisol inhuman peripheral-blood mononuclearcells. International Immunopharmacology1(12), 2109–2115.

Hoffman-Goetz, L. & Zajchowski, S. (1999)In vitro apoptosis of lymphocytes afterexposure to levels of corticosteroneobserved following submaximalexercise. Journal of Sports Medicine andPhysical Fitness 39, 269–274.

Hoffman, T., Hirata, F., Bougnoux, P. et al. (1981) Phospholipid methylationand phospholipase A2 activation incytotoxicity by human natural killercells. Proceedings of the National Academyof Sciences of the United States of America78(6), 3839–3843.

Jiang, C.L., Xu, D., Lu, C.L. et al. (2000)Interleukin-2: structural and biologicalrelatedness to opioid peptides.Neuroimmunomodulation 8(1), 20–24.

Jonsdottir, I.H., Johansson, C., Asea, A. et al.(1996) Chronic intracerebroventricularadministration of β-endorphinaugments natural killer cell cytotoxicityin rats. Regulatory Peptides 62, 113–118.

Jonsdottir, I.H., Hellstrand, K., Thoren, P.& Hoffmann, P. (2000) Enhancement ofnatural immunity seen after voluntaryexercise in rats. Role of central opioidreceptors. Life Sciences 66(13), 1231–1239.

Kay, N., Allen, J. & Morley, J.E. (1984)Endorphins stimulate normal humanperipheral blood lymphocyte naturalkiller activity. Life Sciences 35(1), 53–59.

Kraemer, R.R., Acevedo, E.O.,Dzewaltowski, D. et al. (1996) Effects oflow-volume resistive exercise on β-endorphin and cortisol concentrations.International Journal of Sports Medicine17(1), 12–16.

Kraemer, W.J., Fry, A.C., Warren, B.J. et al.(1992) Acute hormonal responses in elitejunior weightlifters. International Journalof Sports Medicine 13(2), 103–109.

adrenocorticotropin, β-endorphin, β-lipotropin and cortisol to athleticcompetition. Acta Endocrinologica 118(3),332–336.

Rabin, B.S., Moyna, M.N., Kusnecov, A.,Zhou, D. & Shurin, M.R. (1996)Neuroendocrine effect of immunity. In:Exercise and Immune Function (Hoffman-Goetz, L., ed.). CRC Press, Boca Raton,FL: 21–38.

Rahkila, P. & Laatikainen, T. (1992) Effectof oral contraceptives on plasma β-endorphin and corticotropin at rest and during exercise. GynecologicalEndocrinology 6(3), 163–166.

Saland, L.C., van Epps, D.E., Maez, D.,Gutierrez, L. & Samora, A. (1988) Acuteinfusion of chemotactic or enkephalin-analog peptides into rat cerebralventricles: scanning and transmissionelectron microscopy of leukocyteimmigration in vivo. Journal ofNeuroimmunology 18(3), 197–206.

Schmidt, M., Pauels, H.G., Lugering, N. et al. (1999) Glucocorticoids induceapoptosis in human monocytes: potentialrole of IL-1β. Journal of Immunology(Baltimore, Md.) 163(6), 3484–3490.

Smilios I., Pilianidis, T., Karamouzis, M. & Tokmakidis, S.P. (2003) Hormonalresponses after various resistanceexercise protocols. Medicine and Sciencein Sports and Exercise 35(4), 644–654.

Viru, A. & Tendzegolskis, Z. (1995) Plasmaendorphin species during dynamicexercise in humans. Clinical Physiology(Oxford, England) 15(1), 73–79.

Volek, J.S., Kraemer, W.J., Bush, J.A.,Incledon, T. & Boetes, M. (1997)Testosterone and cortisol in relationshipto dietary nutrients and resistanceexercise. Journal of Applied Physiology82(1), 49–54.

Walberg-Rankin, J., Franke, W.D. &Gwazdauska, F.C. (1992) Response of β-endorphin and estradiol to resistanceexercise in females during energybalance and energy restriction.International Journal of Sports Medicine13(7), 542–547.

Weber, R.J. & Pert, A. (1989) Theperiaqueductal gray matter mediatesopiate-induced immunosuppression.Science 14(245), 188–190.

Zitnik, R.J., Whiting, N.L. & Elias, J.A.(1994) Glucocorticoid inhibition ofinterleukin-1-induced interleukin-6production by human lung fibroblasts:evidence for transcriptional and post-transcriptional regulatory mechanisms.American Journal of Respiratory Cell andMolecular Biology 10(6), 643–650.

Kraemer, W.J., Dziados, J.E., Marchitelli,L.J. et al. (1993) Effects of differentheavy-resistance exercise protocols onplasma β-endorphin concentrations.Journal of Applied Physiology 74(1),450–459.

McMurray, R.G., Forsythe, W.A., Mar,M.H. & Hardy C.J. (1987) Exerciseintensity-related responses of β-endorphin and catecholamines. Medicineand Science in Sports and Exercise 19,570–574.

Metzger, J.M. & Stein, E.A. (1984) Beta-endorphin and sprint training. LifeSciences 34, 1541–1547.

Millar, D.B., Hough, C.J., Mazorow, D.L. &Gootenberg, J.E. (1990) Beta-endorphin’smodulation of lymphocyte proliferationis dose, donor, and time dependent.Brain, Behavior, and Immunity 4(3),232–242.

Navolotskaya, E.V., Malkova, N.V.,Zargarova, T.A. et al. (2001) Synthetic β-endorphin-like peptide immunorphinbinds to non-opioid receptors for β-endorphin on T lymphocytes. Peptides22(12), 2009–2013.

Owen, D.L., Morley, J.S., Ensor, D.M. & Miles, J.B. (1998) The C-terminaltetrapeptide of β-endorphin (MPF)enhances lymphocyte proliferativeresponses. Neuropeptides 32(2), 131–139.

Parillo, J.E. & Fauchi, A.S. (1978)Comparison of the effector cells in human spontaneous cellularcytotoxicity: differential sensitivity of effector cells to in vivo and in vitrocorticosteroids. Scandinavian Journal ofImmunology 8, 99–107.

Pasnik, J., Tchorzewski, H., Baj, Z., Luciak,M. & Tchorzewski, M. (1999) Primingeffect of met-enkephalin and β-endorphin on chemiluminescence,chemotaxis and CD11b moleculeexpression on human neutrophils invitro. Immunology Letters 67(2), 77–83.

Pedersen, B.K. & Beyer, J.M. (1986)Characterization of the in vitro effects ofglucocorticosteroids on NK cell activity.Allergy 41(3), 220–224.

Pedersen, B.K. & Hoffman-Goetz, L. (2000) Exercise and the immune system:regulation, integration, and adaptation.Physiology Review 80, 1055–1081.

Pedersen, B.K., Beyer, J.M., Rasmussen, A.et al. (1984) Methylprednisolone pulsetherapy induced fall in natural killer cellactivity in rheumatoid arthritis. ActaPathologica Microbiologica et ImmunologicaScandinavica. Section C 92(5), 319–323.

Petraglia, F., Barletta, C., Facchinetti, F. et al. (1988) Response of circulating

Page 361: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

345

The endocrine and nervous systems work togetherto co-ordinate growth and development, to regulatehomeostasis and to co-ordinate the response of thebody to stress. The study of the interrelationshipbetween these systems is termed neuroendocrino-logy. The immune system is controlled locally bychemical signals generated at the cellular level andsystemically by the neuroendocrine system. How-ever, complex, bi-directional, anatomical and physio-logical interactions exist among the three interrelatedsystems, nervous, endocrine and immune (Masek et al. 2003). All three systems have receptors for ashared set of ligands including cytokines, peptidehormones and neurotransmitters (Haddad et al.2002). Thus, the immune system is capable of exert-ing influence on the neuroendocrine system as well as vice versa. Exercise is a form of stress to thebody, and as such, it elicits the stereotypic, neuro-endocrine stress response first described in 1936 byHans Selye as ‘the general adaptation syndrome’(Selye 1936, p. 32). The complexity of the neuro-endocrine immune system is such that the immuneresponse to exercise varies according to intensityand duration of exercise, environmental conditions,nutritional factors, level of recovery from previoustraining and tissue damage (Nieman 1997; Pedersen& Hoffman-Goetz 2000).

The aim of this chapter is to examine the neuro-endocrine immune response to exercise stress fromseveral perspectives within the context of: (i) acuteexercise; (ii) exercise training; and (iii) exercise-induced muscle damage. The key immune sys-tem components are described in Table 25.1. Theimmune response to acute exercise is influenced by

intensity and duration of exercise, the recovery state of the body and the availability of nutrientsduring the exercise. In many respects, the responsesand adaptations to exercise training are the cumu-lative influence of repeated acute exercise bouts andresources provided to the body for recovery andadaptation. Exercise-induced muscle damage activ-ates the immune system both locally and systemic-ally and provides a model to study the inflammatoryarm of the immune system. The neuroendocrineimmune response to muscle damage and otherstresses to muscle provides insight into the role that this complex system may play in eliciting adaptations to exercise training, for example musclehypertrophy. Understanding the implications ofexercise-induced immune modulations and under-standing that the immune system may have a role in producing physiological adaptations to exerciseis important to the design of exercise programs forhealth and for athletic performance.

Sympathetic nervous system

The autonomic nervous system includes the para-sympathetic nervous system that controls restingfunctions and the sympathetic nervous system thatenables the body to become physically active, as inthe ‘fight-or-flight’ response. Responses to stress,regardless of the nature of the stress, are co-ordinated collectively by the sympathetic nervoussystem and the hypothalamic–pituitary–adrenal(HPA) axis (discussed subsequently) (Tsigos &Chrousos 2002). Epinephrine and norepinephrine,also known as adrenaline and noradrenaline or

Chapter 25

Neuroendocrine Modulation of the Immune Systemwith Exercise and Muscle Damage

MARY P. MILES

Page 362: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

346 chapter 25

Tab

le 2

5.1

Key

com

pone

nts

of th

e im

mun

e sy

stem

. (D

ata

from

Lile

s &

Van

Voo

rhis

199

5; S

heph

ard

199

7; E

lenk

ov et

al.

2000

; Riv

est 2

001;

Suz

uki e

t al.

2002

;St

eens

berg

et a

l. 20

03.)

Com

pone

ntPr

oduc

tion

/loc

atio

nK

ey fu

ncti

ons

Leuk

ocyt

esN

eutr

ophi

lsPr

oduc

ed in

bon

e m

arro

w a

nd re

leas

ed to

the

circ

ulat

ion.

In

flam

mat

ion

and

nat

ural

imm

une

def

ense

aga

inst

infe

ctio

n.

Man

y ad

here

to v

ascu

lar e

ndot

helia

l cel

ls, p

arti

cula

rly

in

Prod

uce

free

rad

ical

oxy

gen

spec

ies

that

des

troy

bac

teri

a th

e lu

ngs

and

inju

re n

earb

y ce

lls. R

emov

e sm

all d

ebri

s in

the

area

of

infe

ctio

n or

infl

amm

atio

n by

the

proc

ess

of p

hago

cyto

sis

Mon

ocyt

es/m

acro

phag

esM

onoc

ytes

are

pro

duc

ed in

bon

e m

arro

w a

nd a

re fo

und

in

Nat

ural

imm

unit

y pr

otec

tion

aga

inst

vir

al in

fect

ion

and

tum

ors,

the

bloo

d. A

fter

leav

ing

the

circ

ulat

ion,

mon

ocyt

es

phag

ocyt

osis

of c

ellu

lar d

ebri

s, p

rod

ucti

on o

f infl

amm

ator

yd

iffe

rent

iate

to b

ecom

e m

acro

phag

es

cyto

kine

s (T

NF-

α, IL

-1β,

IL-6

, IL

-10,

IL-1

2). F

ollo

win

g ph

agoc

ytos

is a

nd a

ctiv

atio

n, m

acro

phag

es a

re a

ble

to

‘pre

sent

’ ant

igen

s to

T-l

ymph

ocyt

es to

act

ivat

e an

tige

n-d

epen

den

t/ac

quir

ed im

mun

e d

efen

ses

Lym

phoc

ytes

Nat

ural

kill

er (N

K)

Prod

ucti

on b

y bo

ne m

arro

w, f

ound

in b

lood

and

att

ache

d

Nat

ural

imm

unit

y vi

a no

n-m

ajor

his

toco

mpa

tibi

lity

com

plex

ce

lls (C

D3–

CD

16+ 5

6+ )to

vas

cula

r end

othe

lial c

ells

in ly

mph

oid

tiss

ues.

(MH

C)-

rest

rict

ed c

ytot

oxic

ity,

e.g

. kill

ing

vira

lly in

fect

ed c

ells

an

d s

ome

tum

or c

ells

. Im

port

ant f

or e

arly

def

ense

aga

inst

vi

ruse

s an

d s

ome

mal

igna

ncie

s

Cyt

otox

ic T

-lym

phoc

ytes

Prod

ucti

on b

y bo

ne m

arro

w a

nd m

atur

atio

n in

the

thym

us

MH

C-r

estr

icte

d c

ytot

oxic

ity.

Impo

rtan

t for

cel

l-m

edia

ted

,(C

D3+

CD

8+ )gl

and

. Mat

ure

cells

foun

d in

lym

phat

ic ti

ssue

, spl

een

‘acq

uire

d’ i

mm

une

resp

onse

s to

kill

infe

cted

cel

lsan

d b

lood

Hel

per T

-lym

phoc

ytes

Prod

ucti

on b

y bo

ne m

arro

w a

nd m

atur

atio

n in

the

thym

us

Co-

ord

inat

ion

of im

mun

e re

spon

ses.

Und

iffe

rent

iate

d (T

h0)

(CD

3+C

D4+ )

glan

d. M

atur

e ce

lls fo

und

in ly

mph

atic

tiss

ue, s

plee

n C

D4+

cells

will

be

acti

vate

d fo

r dif

fere

ntia

tion

to e

ithe

r the

an

d b

lood

Th1

CD

4+ce

lls th

at re

gula

te c

ellu

lar i

mm

unit

y or

the

Th2

C

D4+

cells

that

regu

late

hum

oral

imm

unit

y an

d s

ome

infl

amm

ator

y fu

ncti

ons.

A s

mal

l min

orit

y of

CD

4+ce

lls

prod

uce

TG

F-β

and

are

des

igna

ted

Th3

CD

4+ce

lls

B-l

ymph

ocyt

es (C

D19

+ )Pr

oduc

ed b

y bo

ne m

arro

w, u

pon

acti

vati

on b

y an

tige

n th

ey

Stim

ulat

ion

by a

ntig

en a

nd c

ytok

ines

from

Th1

type

CD

4+

dif

fere

ntia

te to

bec

ome

plas

ma

cells

. Mat

ure

B-c

ells

foun

dT

-lym

phoc

ytes

ind

uces

pro

duc

tion

of i

mm

unog

lobu

lins

in m

any

extr

acel

lula

r flui

ds

incl

udin

g bl

ood

and

muc

ous,

(a

ntib

odie

s)an

d s

tore

d in

lym

phat

ic ti

ssue

s

Imm

unog

lobu

lins

(Ig)

Prod

uced

by

plas

ma

cells

(B-c

ells

act

ivat

ed fo

r ant

igen

B

ind

to m

olec

ular

and

cel

lula

r ant

igen

s (p

arti

cula

rly

bact

eria

l)

prod

ucti

on) a

nd re

leas

ed. F

ound

in b

lood

, sal

iva,

muc

osal

to

form

an

anti

bod

y-an

tige

n co

mpl

ex th

at in

duc

es p

hago

cyto

sis

secr

etio

ns a

nd th

roug

hout

the

bod

yby

neu

trop

hils

and

mac

roph

ages

to e

limin

ate

the

anti

gen

Page 363: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 347

Cyt

okin

esIn

terf

eron

-γT

h0 a

nd T

h1 C

D4+

T-c

ells

, CD

8+T

-cel

ls (T

c1 s

ubse

t),

Stim

ulat

es m

acro

phag

e ac

tiva

tion

, neu

trop

hils

and

NK

cel

ls(I

FN-γ

)N

K c

ells

and

pro

duc

tion

of a

ntib

odie

s by

B-c

ells

; inh

ibit

s T

h2 c

ytok

ine

prod

ucti

on b

y C

D4+

T-c

ells

Tum

or n

ecro

sis

fact

or-α

Prod

uced

by

mon

ocyt

es/m

acro

phag

es a

nd N

K c

ells

and

to

Ant

i-tu

mor

act

ivit

y, in

itia

tion

of i

nflam

mat

ion

via

chem

okin

e(T

NF-

α)a

less

er e

xten

t by

neut

roph

ils, T

- and

B-l

ymph

ocyt

es a

nd

ind

ucti

on, r

ecru

itm

ent o

f neu

trop

hils

and

mon

ocyt

es a

nd

othe

r cel

lsin

duc

tion

of I

L-6

syn

thes

is

Inte

rleu

kin-

1β(I

L-1

β)Pr

oduc

ed b

y m

onoc

ytes

/mac

roph

ages

Ind

ucti

on o

f cer

ebra

l res

pons

e to

infl

amm

atio

n, e

.g. f

ever

, st

imul

atio

n of

pro

stag

land

in E

2pr

oduc

tion

, sti

mul

atio

n of

IL

-2 re

cept

or e

xpre

ssio

n, in

duc

es IL

-6 s

ynth

esis

Inte

rleu

kin-

2 T

h0 a

nd T

h1 C

D4+

T-c

ells

, CD

8+T

-cel

ls (T

c1 s

ubse

t)T

h1 c

ytok

ine,

pot

ent s

tim

ulat

ion

of N

K c

ell a

ctiv

ity,

sti

mul

atio

n(I

L-2

)of

lym

phoc

yte

prol

ifer

atio

n an

d a

ntib

ody

secr

etio

n by

B-c

ells

Inte

rleu

kin-

4T

h0 a

nd T

h2 C

D4+

T-c

ells

, B-c

ells

Stim

ulat

ion

of T

h2 c

ells

, sti

mul

atio

n of

imm

unog

lobu

lin(I

L-4

)pr

oduc

tion

and

pro

lifer

atio

n by

B c

ells

, sti

mul

atio

n of

alle

rgic

resp

onse

s vi

a Ig

E p

rod

ucti

on, i

nhib

its

cyto

kine

pro

duc

tion

by

Th1

CD

4+ce

lls

Inte

rleu

kin-

6 T

h0 a

nd T

h1 C

D4+

T-c

ells

, CD

8+T

-cel

ls (T

c1 s

ubse

t) a

nd

Th2

cyt

okin

e, s

tim

ulat

es B

- and

cyt

otox

ic T

-cel

ls, i

nduc

tion

of

(IL

-6)

mon

ocyt

es/m

acro

phag

es, b

ut n

earl

y al

l cel

ls c

an b

e IL

-2 p

rod

ucti

on, s

tim

ulat

ion

of a

cute

pha

se p

rote

in s

ynth

esis

,in

duc

ed to

pro

duc

e IL

-6, m

ost n

otab

ly m

uscl

e ce

llsac

tiva

tion

of t

he H

PA a

xis,

inhi

biti

on o

f TN

F-α

and

IL-1

βsy

nthe

sis,

sti

mul

atio

n of

IL-1

0 an

d IL

-1ra

syn

thes

is

Inte

rleu

kin-

8Pr

oduc

ed b

y m

onoc

ytes

, mac

roph

ages

and

end

othe

lial c

ells

Che

mok

ine,

recr

uits

neu

trop

hils

to in

flam

mat

ory

site

s,

(IL

-8)

stim

ulat

es p

rod

ucti

on o

f rea

ctiv

e ox

ygen

spe

cies

and

d

egra

nula

tion

by

neut

roph

ils

Inte

rleu

kin-

10Pr

oduc

ed b

y T

h0 a

nd T

h2 C

D4+

T-c

ells

, mon

ocyt

es a

nd

Th2

cyt

okin

e, in

hibi

ts c

ytok

ine

prod

ucti

on b

y T

h1 C

D4+

T-c

ells

,(I

L-1

0)B

-cel

ls, a

s w

ell a

s pi

tuit

ary

and

hyp

otha

lam

ic c

ells

mon

ocyt

es a

nd m

acro

phag

es, s

tim

ulat

es B

-lym

phoc

yte

prol

ifer

atio

n an

d a

ntib

ody

secr

etio

n

Inte

rleu

kin-

12Pr

oduc

ed b

y m

onoc

ytes

Stim

ulat

es th

e T

h1 im

mun

e pa

thw

ay, p

rom

otes

cyt

otox

ic

(IL

-12)

acti

vity

of C

D8+

T-l

ymph

ocyt

e an

d N

K c

ells

, inh

ibit

s Ig

Ese

cret

ion

from

B-l

ymph

ocyt

es

Tra

nsfo

rmin

g gr

owth

Th3

CD

4+T

-cel

ls, m

acro

phag

es a

nd o

ther

cel

lsIn

hibi

tion

of N

K c

ell a

ctiv

ity,

B- a

nd T

-cel

l pro

lifer

atio

n an

dfa

ctor

β(T

GF-

β)so

me

mac

roph

age

func

tion

s; s

tim

ulat

ion

of Ig

A s

ecre

tion

by

B-l

ymph

ocyt

es

Page 364: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

348 chapter 25

simply catecholamines, are the neurotransmittersreleased by the sympathetic nervous system. Ac-tivation of the sympathetic nervous system via thelocus ceruleus–norepinephrine system (LC/NE)stimulates release of epinephrine from the adrenalmedulla and norepinephrine from axon terminals of sympathetic neurons. Both of these neurotrans-mitters increase in the bloodstream during exercise,although the relative concentrations of norepine-phrine exceed those of epinephrine by severalorders of magnitude (Weicker & Werle 1991; Kjær & Dela 1996). There is a linear increase in the con-centration of catecholamines as the duration of exer-cise increases (Kjær & Dela 1996). In contrast, theincrease in catecholamines in response to increas-ing exercise intensity is of a greater magnitude,more closely approximating an exponential increase(Kjær & Dela 1996).

Leukocyte trafficking

The most dramatic effect of catecholamines on theimmune system is to draw leukocytes from extra-vascular storage depots to the circulation. Researchstudies involving infusion of epinephrine or nore-pinephrine to match exercise levels or involvingblockade of catecholamine receptors (adrenergicreceptors) during exercise clearly indicate thatepinephrine (ligand for β1- and β2-adrenergic recep-tors) recruits lymphocytes and neutrophils to thecirculation during exercise (van Titts et al. 1990;Kappel et al. 1991; Benschop et al. 1994; Schedlowskiet al. 1996). Norepinephrine (strong β1- and weak β2-adrenergic receptor ligand) has a smaller effecton circulating leukocytes than epinephrine. Thus, itgenerally is accepted that increased intracellularcyclic adenosine monophosphate (cAMP) inducedby epinephrine binding to β2-adrenergic receptors is a primary stimulus driving lymphocytes and neutrophils into the circulation (Boxer et al. 1980;Weicker & Werle 1991; Schedlowski et al. 1996).

Tissues where immune cells are produced orreside, including the thymus, spleen, lymph nodes,tonsils, bone marrow and gut-associated lymphoidtissue (GALT), are innervated by the sympatheticnervous system via noradrenergic and or neuropep-tide Y (NPY) nerve terminals (Elenkov et al. 2000).

This ‘hard-wire’ connection has a major role in func-tional modulation of immune cells, but it has littleeffect on exercise-induced leukocyte trafficking.

Leukocyte function

The view that catecholamines have an immuno-suppressive net effect is losing favor to a view thatrecognizes a more complex variety of responses(Elenkov et al. 2000). As illustrated in Fig. 25.1, theacute influence of catecholamines on the immunesystem is complex, but generally leads to suppres-sion of the Th1 system (production of interleukin-2[IL-2], interferon-γ [IFN-γ] and regulation of cellu-lar immunity), no direct effect on the Th2 system(production of IL-4, IL-5, IL-6 and IL-10, and regula-tion of humoral immunity) and mixed effects on theinflammatory system. A portion of the proinflam-matory response to catecholamines is the result ofdisinhibition of inflammatory cytokine synthesisthat occurs when catecholamines inhibit IL-2 andIL-12 synthesis, i.e. stimulation via removal of inhibition. In contrast to acute exercise, chronicexposure to catecholamines results in desensitiza-tion and a cell type-specific down-regulation of β2-adrenergic receptors or other components of cellsignaling (Elenkov et al. 2000). Thus, acute andchronic stresses may have divergent effects on theimmune system.

Catecholamines also are capable of modulatingnatural killer (NK) cell function. Both epinephrineand norepinephrine, but particularly epinephrine,have induced increases in circulating NK cells anddecreases in the per cell cytotoxic activity of thosecells (Schedlowski et al. 1993; Klokker et al. 1997;Kappel et al. 1998). The decrease in activity is likelyto be a function of catecholamine-induced decreasesin IL-2 and IL-12, cytokines that up-regulate NK-cellcytotoxicity (see Fig. 25.1).

While many cellular responses are induced dir-ectly by rises in intracellular cAMP (Borger et al.1998), it has been suggested that the functional modulations of lymphocytes in response to cate-cholamine increases are mediated by macrophagesand nitric oxide (Rabin et al. 1996). Evidence for thismechanism has also been presented in the rodentmodel (Blank et al. 1997). An example relevant to

Page 365: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 349

NEE

STRESS

CNS(+)

TNF-αIFN-γ

IL-12IL-2

IL-8GM-CSF

IL-10MIP-1α

IL-6TGF-β

Acute phase response

Th1 responses:

Hypothalamus LC/NE(+)

(+)

(+)

Spinal cord

Other

Vascular tissue

Lymphatic tissue

MyocardiumBonemarrow

Sympathetic nerve terminals

Pituitary

NE/NPY

Adrenal gland

Circulation

E/NE β-adrenergic stimulation

MedullaCortex

IL-4

Th2 responses:

Leukocyte demargination

Neutrophil phagocytosis and oxidative burst

Natural killer cell cytolytic activity

B-cell Ig production/secretion

Immune cell responses: Inflammatory responses:

Fig. 25.1 (+) Indicates stimulation; dashed arrows indicate effect induced by removal of inhibition; boxes indicateneurotransmitters. CNS, central nervous system; E, epinephrine; GM-CSF, granulocyte-macrophage colony-stimulatingfactor; Ig, immunoglobulin; IL, interleukin; LC/NE, locus ceruleus–norepinephrine; NE, norepinephrine; NPY,neuropeptide Y; TGF, transforming growth factor; TNF, tumor necrosis factor. (Data from Sanders et al. 1997; Borger et al.1998; Elenkov et al. 2000; Kohm & Sanders 2001.)

Page 366: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

350 chapter 25

acute exercise is that Kappel et al. (1991) were able todemonstrate that NK-cell activity per cell was lower2 h following epinephrine infusion. There was atwofold increase in monocytes at this time, and itwas hypothesized that prostaglandins produced bythe monocytes inhibited the cytolytic activity of theNK cells. As hypothesized, when prostaglandinproduction by monocytes was blocked using indo-methacin, the per cell decrease in NK-cell activityalso was blocked. This suggests that prostaglandinsproduced by monocytes, rather than a direct effectof epinephrine, inhibited NK-cell function. By con-trast, it should be noted that Nieman et al. (1995a)measured decreased NK-cell activity despite indo-methacin administration. Thus, this mechanismmay not be consistent across all situations.

Hypothalamic–pituitary–adrenal axis

The HPA axis consists of signals feeding forwardfrom the hypothalamus to the anterior pituitary andthen to the adrenal cortex (Tsigos & Chrousos 2002;Beishuizen & Thijs 2003). In response to many formsof stress, the hypothalamus releases corticotropin-releasing hormone (CRH) and vasopressin (AVP).CRP, and to a lesser extent AVP, stimulate the anter-ior pituitary to produce adrenocorticotropic hor-mone (ACTH). The main role of AVP is to promotefluid resorption by the kidneys. As an endocrinehormone, ACTH travels through the circulation tostimulate the adrenal cortex to release glucocor-ticoid hormones, most notably cortisol. As such, cortisol is the final product of the HPA axis. Theeffects of cortisol on the immune system and thedetails of the relationships amongst the HPA axiscomponents and the inflammatory cytokines areillustrated in Fig. 25.2. Cortisol acts in a negativefeedback fashion to inhibit CRH and ACTH release.As a steroid hormone, cortisol is able to diffusethrough plasma membranes and bind to intracel-lular receptors (Riccardi et al. 2002). The cortisol/glucocorticoid receptor complex affects cellularfunction primarily by up-regulating and down-regulating transcription of various proteins, but alsovia more rapid means such as a Ca2+-dependentmechanism (Buckingham et al. 1996). Cortisol sup-

presses a number of immune responses and is a keyregulatory element preventing the immune systemfrom over responding to immune challenges andbecoming destructive. For example, inflammationleft unchecked will lead to excessive tissue destruc-tion and possibly death (Northoff et al. 1995; Suzukiet al. 2002).

The effects of cortisol on the immune system areconsistent with down-regulation of immune func-tion, particularly down-regulation of inflammatoryfunctions. For this reason, glucocorticoid analogsare commonly used to treat inflammatory and auto-immune pathologies (Ashwell et al. 2000). Bindingof cortisol to the intracellular glucocorticoid recep-tor leads to activation of a glucocorticoid responseelement (GRE) and an array of transcriptionalresponses (Pitzalis et al. 2002). Of particular interestare the up-regulation of annexin I (formerly referredto lipocortin-1) and anti-inflammatory proteins, forexample IL-1 receptor antagonist, and the down-regulation of cell adhesion molecules (CAM) andproinflammatory cytokines (Levine et al. 1996;Pitzalis et al. 2002).

The level of HPA activity ideally falls within a range that allows for mounting an effectiveimmune/inflammatory response when needed, butdoes not allow excessive immune activity to becomedestructive. Excessive stress, in a variety forms, may result in chronically elevated cortisol levelsand induce immunosuppression (Buckingham et al.1996). For example, immunosuppression associ-ated with depression has been linked to elevationsin cortisol (Leonard & Song 1996). Reciprocally,adrenocortical insufficiency associated with lowglucocorticoid release has been associated with in-creased susceptibility to autoimmune/inflammatoryconditions (Buckingham et al. 1996). Pathologicaldisorders that result in over and underproductionof glucocorticoids are identified as Cushing’s andAddison’s diseases, respectively. Within the non-pathological spectrum of adrenocortical reactivity,high and low stress responders have been identifiedwith respect to the release of ACTH in response tostress (Petrides et al. 1997; Deuster et al. 1999). Thus,variability in the magnitude of the HPA response tostress is expected and a number of factors modulate

Page 367: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 351

Infection/tissue trauma STRESS

CNS

CRH

HypothalamusHypothalamic

Pituitary

Gonadal

Axis

Hypothalamic

Pituitary

Thyroid

Axis

(+)

(+)

(–)

(–)

(–)

(+)

(+)

Medulla

TNF-αIL-1βIL-6LIF

TNF-αIL-1βIL-6

IL-2

Th1 responses:

T Lymphocyte proliferation

Neutrophil release from bone marrow

Lymphocyte entry into tissues

B cell IgE secretion

Monocyte/macrophage activation

ACTH

Adrenal gland

Circulation

Pituitary

Cortisol

Cortex

Circulation

IL-4

Th2 responses:

IL-10

Immune cell responses:

Fig. 25.2 (+) Indicates stimulation; (–) indicates inhibition; boxes indicate neurotransmitters or hormones. ACTH,adrenocorticotropic hormone; CNS, central nervous system; CRH, corticotropin-releasing hormone; E, epinephrine; IL, interleukin; LC/NE, locus ceruleus-norepinephrine; LIF, leukemia inhibitory factor; NE, norepinephrine; NPY,neuropeptide Y; TGF, transforming growth factor; TNF, tumor necrosis factor. (Data from Weicker & Werle 1991;Chesnokova & Melmed 2002; Haddad et al. 2002; Riccardi et al. 2002; Tsigos et al. 2002.)

Page 368: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

352 chapter 25

this variability. This variability also will manifest invariability in the immune response to stress, regard-less of the stressor.

The two-way communication between the im-mune and neuroendocrine systems is such thatinflammatory cytokines, particularly tumor nec-rosis factor-α (TNF-α), IL-1β, IL-6 and leukemiainhibitory factor (LIF) can stimulate the HPA axis toinduce the release of cortisol (Mastorakos et al. 1993;Chesnokova & Melmed 2002; Tsigos & Chrousos2002). LIF is necessary for inflammation-inducedrelease of ACTH and cortisol, and may be a poten-tiator of TNF-α and IL-1β induced activation of the HPA axis (Chesnokova & Melmed 2000, 2002;Chesnokova et al. 2002). That is, the chemical signalsthat promote the inflammatory process also initiatea negative feedback loop to turn their own activitydown. The reverse of this relationship has also been demonstrated. Specifically, most pituitary hor-mones can be produced by lymphocytes (Carr &Blalock 1990). For example, lymphocytes stimulatedwith recall antigen or with IL-12 will producegrowth hormone (Malarkey et al. 2002).

In addition to modulating the immune system,glucocorticoids play an important role in enablingthe body to respond to stress in such a way thatincreases the likelihood of survival in stressful situations. Glucocorticoids inhibit release of sexsteroids and growth hormone (Tsigos & Chrousos2002), thus removing the likelihood of the need toallocate resources of the body to non-essentialgrowth. Further, glucocorticoids inhibit thehypothalamic–pituitary–thyroid axis, an effect thatresults in a lowering of basal metabolic rate (Tsigos& Chrousos 2002). To accommodate the need forenergy in the tissues forced to respond to stress, glucocorticoids promote gluconeogenesis, glyco-genolysis, lipolysis and proteolysis (McMurray &Hackney 2000; Steinacker et al. 2004). By promotinggluconeogenesis and glycogenolysis by the liver,cortisol helps to maintain blood glucose levels and it is considered to be a ‘glucoregulatory’ hormone.Thus, growth processes are turned off, fertility isdecreased, metabolic rate is turned down to decreasethe overall demand for energy on the body, andenergy substrates stored for a ‘rainy day’ are madeavailable to the body.

Leukocyte trafficking

The effects of glucocorticoids on leukocyte migra-tion from tissue to tissue occur in a delayed timecourse relative to catecholamines. Shifts in leuko-cytes typically peak approximately 4 h after eleva-tions in cortisol are induced (Rabin et al. 1996).Broken down by leukocyte type in the circulation,the response to glucocorticoid increases are concen-tration decreases in lymphocytes and monocytesand increases in neutrophils (Rabin et al. 1996;Nieman 1997). The net effect is a rise in leukocytecount exclusively owing to an influx of neutrophils.Pharmacological analogs also produce this profile.

Cytokines

Glucocorticoids are capable of inhibiting produc-tion of many cytokines, including IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, granulocyte-macrophage colony-stimulating factor (GM-CSF),TNF-α and IFN-α (Ashwell et al. 2000; Riccardi et al.2002). The influence on Th1 cytokine production is greater than that for Th2 cytokine production; for example, IL-12 synthesis is profoundly inhibitedand IL-10 synthesis is only moderately inhibited(Ashwell et al. 2000). As such, glucocorticoids areconsidered potent inhibitors of cellular immunityand inflammation.

Leukocyte function

The functional consequences of these glucocorticoid-induced genomic and non-genomic effects are wide-spread through the immune system. Both NK- andT-cell functions are inhibited by cortisol (Ramirez &Silva 1997; Zhou et al. 1997; Ashwell et al. 2000). Theability of NK cells to lyse target cells is used to de-termine NK cell activity (NKCA). Cortisol down-regulates NKCA by decreasing synthesis of effectorproteins (Zhou et al. 1997). The cellular immuneresponse is dependent on the clonal proliferation of antigen-specific T- and B-cells. Lymphocyte pro-liferation assays are a popular functional meas-urement performed in vitro by exposing these cellsto cytokines or polyclonal mitogens or antigens toinduce proliferation. T-cell proliferation is inhibited

Page 369: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 353

by cortisol (Ashwell et al. 2000; Cancedda et al. 2002).The effect on B-cells is mixed, with inhibition ofimmunoglobulin G (IgG) and IgA and stimulationof IgE synthesis (Ashwell et al. 2000).

Acute exercise

While it is important to understand the effects of thesympathetic nervous system and HPA axis hor-mones in isolation, exercise-induced immunomo-dulation is the result of the collective influences ofmany physiological responses occurring simultan-eously or in sequence. Owing to the complexity of the systems involved and the number of potentialfactors that can enhance or attenuate the stress to thesystem as a whole, the array of potential influenceson the immune response is virtually infinite. A single bout of exercise can induce the immune sys-tem to transiently redistribute leukocytes to thebloodstream and amongst tissues, alter the func-tional capacity of leukocytes, induce the release of ahost of molecules that regulate immune function,produce transient or prolonged inflammation, andin doing so, change the overall immune defenselevel that the body has for protection against infec-tion and tumor cells. The extent of these changestypically is greater as the magnitude of the exercisestress increases. The typical determinants of stressmagnitude are intensity and duration of exercise(McMurray & Hackney 2000). However, the level ofstress to which the neuroendocrine immune systemmust respond is enhanced by additional factorssuch as insufficient recovery from previous exercise(Ronsen et al. 2002a, 2002b; McFarlin et al. 2003), lowcarbohydrate availability (Nieman et al. 1998; Greenet al. 2003), hypoxic conditions (Klokker et al. 1995;Niess et al. 2003) and heat stress (Brenner et al. 1998;Mitchell et al. 2002).

Leukocyte trafficking

Activation of the sympathetic nervous system elicitsthe exercise-induced increases among the differentleukocyte subsets. While there is tremendous inter-individual variability in the magnitude of response,the overall pattern of leukocyte redistribution dur-ing exercise is robust and consistent (see Shephard

1997 for review). NK cells have the greatest β2-adrenergic receptor density, followed by CD8+ T-lymphocytes and B-lymphocytes and monocytes,and then by T-helper type 1 CD4+ T-lymphocytes(Maisel et al. 1990; Schedlowski et al. 1996). T-helpertype 2 CD4+ T-lymphocytes are the only lymphocytesubset that does not express β-adrenergic recep-tors (Sanders et al. 1997). Consistent with the cate-cholamine response, the NK response to exerciseincreases with intensity and duration of exerciseand the reported increases during exercise range allthe way from 50% to 900% (Shephard 1997). Theremaining lymphocyte subsets respond less dram-atically as the density of β-adrenergic receptorsdecreases. Additionally, the cells that enter the circulation are predominately memory/activatedcells with short telomeres in the case of lymphocytes(Bruunsgaard et al. 1999; Pedersen & Hoffman-Goetz 2000), and segmented rather than bandednuclei in the case of neutrophils (Miles et al. 1998).This indicates that the influx of cells to the circula-tion is made up of mature cells, rather than of newlyproduced or released immature/naïve cells.

Cortisol gains increasingly more influence overthat of the catecholamines as the duration of exer-cise continues. Approximately 1.5 h into enduranceexercise, the influence of cortisol on leukocyte distribution and function overtakes that of the catecholamines (Nieman 1997). If the duration and intensity of exercise are sufficient to create anincrease in cortisol, then a cortisol-driven lympho-cytosis occurs for 1–3 or 4 h post-exercise (Nieman1997).

Neutrophils are similar to lymphocytes in thatthey increase in the circulation during exercise in adose–response fashion, and they differ from lym-phocytes in that they have a second wave of eleva-tion that is delayed and sustained in the circulationa few hours following exercise that induces a sig-nificant cortisol increase (Peake 2002). While epine-phrine is credited with a portion of the inducedincrease during exercise, a number of additional factors have been associated with the rise in neutro-phils including IL-6, IL-8, granulocyte-colony stimu-lating factor (G-CSF), growth hormone and muscledamage (Hetherington & Quie 1985; Suzuki et al.1999; Peake 2002; Yamada et al. 2002). The delayed

Page 370: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

354 chapter 25

elevation of neutrophils appears to be the effect ofcortisol and includes the entry of both immatureband neutrophils from the bone marrow and maturesegmented cells from marginal pools (Suzuki et al.1996, 1999).

Monocytes increase in the circulation as part ofthe transient response to acute exercise (Woods &Davis 1994). This response is likely to be induced byenhanced hemodynamic forces and catecholamine-induced decreases in adhesion to vascular endo-thelial cells (Woods et al. 1999).

Cytokines

The pattern of cytokine production in response toexercise is not entirely consistent with the catechola-mine and glucocorticoid response. That is, stimula-tion of the sympathetic nervous system shouldinhibit Th1 cytokine production (TNF-α, IL-2, IL-12and IFN-γ), have little effect on Th2 cytokine pro-duction (IL-4), and stimulate inflammatory cytokineproduction (IL-6, IL-8, IL-10, transforming growthfactor-β [TGF-β]). Activation of the HPA axis shouldinhibit production of some Th1 cytokines (IL-2) andsome inflammatory cytokines (TNF-α, IL-1β, IL-6),and stimulate production of some Th2 cytokines(IL-4 and IL-10). These responses are what would beexpected if catecholamines and glucocorticoids con-trolled the cytokine response to exercise. Consistentwith this model is the tendency for IL-2 and IFN-γproduction not to change or to decrease in responseto exercise (Shephard 1997; Ibfelt et al. 2002; Suzukiet al. 2002). Also consistent with this model is thetendency for IL-8 and IL-10 to increase in responseto exercise (Shephard 1997; Nieman et al. 2001;Pedersen et al. 2001; Suzuki et al. 2002). However,exercise elicits an inflammatory or ‘inflammatory-like’ response in a dose-dependent fashion thatincludes elevations in TNF-α, IL-1β, IL-6, IL-10 andIL-1 receptor antagonist (IL-1ra) (Ostrowski et al.2000; Pedersen et al. 2001; Suzuki et al. 2002). Thus,the exercise-induced increase in cortisol occurs inpart because the rise in inflammatory cytokinesstimulates the HPA axis via the two-way com-munication network between the immune and neuroendocrine systems (Smith, L.L. & Miles 2000;Steensberg et al. 2003).

The single most responsive cytokine to exercise is IL-6 which increases exponentially as the dura-tion of exercise progresses (Pedersen et al. 2001) andperhaps in correlation with rises in epinephrine(Papanicolaou et al. 1996). However, factors otherthan epinephrine elicit the vast majority of the exercise-induced rise in IL-6 (Jonsdottir et al. 2000;Steensberg et al. 2001). With respect to magnitudeand time course, IL-6 concentrations increase fromtwo to 100-fold in the blood during endurance exercise and typically return to resting levels withina few hours post-exercise. When exercise is ofshorter duration and biased toward the develop-ment of high eccentric forces within active muscles,the kinetics are delayed and a relatively small peakof IL-6 has been measured a few hours post-exercise(Bruunsgaard et al. 1997a).

A key distinction to be made regarding IL-6 is that, while it has a role in the inflammatoryresponse, it is produced more readily by skeletalmuscle during exercise than by leukocytes, adipose,or the liver (Jonsdottir et al. 2000; Pedersen et al.2001; Febbraio et al. 2003b). A number of physiolo-gical roles for IL-6 in the response to exercise havebeen identified in recent years, and the regulatoryrole of IL-6 beyond the inflammatory response is an emerging area of research (MacDonald et al. 2003; Steensberg et al. 2003). For example, as muscle glycogen is depleted, AMP-activated pro-tein kinase (AMPK) increases, and a strong correla-tion between AMPK and IL-6 release has beenmeasured (MacDonald et al. 2003). Thus, IL-6 maybe involved in systemic signaling related to energyneeds of muscle cells during exercise. Plasma IL-6increases occur in the absence of muscle damageand are independent of other markers of inflam-mation. Exercising, but not resting, skeletal musclecells release IL-6 (Febbraio et al. 2003b). Further, IL-6 gene expression (IL-6 mRNA) was detected in muscle, but not in mononuclear leukocytes(includes monocytes) collected from the blood fol-lowing strenuous exercise (Ostrowski et al. 1998b).Thus, IL-6 will increase in the absence of tissue damage and should be considered both as part of the inflammatory response and as part of aninflammatory-independent response to exercise(Pedersen et al. 2001).

Page 371: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 355

Leukocyte function

Intensity and duration of exercise play an importantrole in the subsequent response of immune cells,including neutrophils. The effects of exercise aresomewhat consistent in that exercise that is moder-ate in duration and intensity transiently enhancesfunctions such as phagocytosis, degranulation ofproteolytic enzymes and oxidative burst (Pyne1994). If exercise is of sufficient intensity and orduration to elicit an increase in circulating gluco-corticoids, then there are a number of functionalconsequences to cells of the immune system. Theoxidative burst of neutrophils tends to be enhancedby exercise (Pyne 1994; Suzuki et al. 1996, 1999).There is some evidence that exercise-induced in-creases in IL-6, growth hormone and glutathionemay be responsible for priming circulating neutro-phils (Atalay et al. 1996; Suzuki et al. 1999). If theexercise lasts for a prolonged period of time or isextremely strenuous, for example a marathon, thenthe trend is for these functions to decrease (Müns1993; Pyne 1994). This decrease may be a function ofthe diminished capacity of neutrophils to respondto stimuli after they have already been activated(Pyne 1994). Thus, moderate exercise may enhanceand strenuous or intense exercise may reduce thefirst line of defense against bacterial and viral infections.

Exercise of a variety of intensities and durationshas been measured to enhance several macrophagefunctions (Woods et al. 1994, 1999). Chemotaxis isthe process by which monocytes and other immunecells follow a chemical gradient to move towardsites of inflammation or infection. Phagocytosis andelements of the associated oxidative burst also arestimulated by intense exercise and mediated by corticosteroids (Ortega et al. 1996). While the influ-ence of corticosteroids is anti-inflammatory, the neteffect of down-regulation of various inflammatorystimuli may be an increase in macrophage activa-tion and activity (Woods et al. 1994; Ortega et al.1997). However, Woods et al. (1994) have demon-strated in a murine model that the influence may beindirect and related to reactive nitrogen molecules,such as nitric oxide. Thus, the ability of monocytesand macrophages to response to injury or infection

is likely to be improved for a period of time afterexercise.

One factor attenuating the functional effects ofcortisol on immune function is that the sensitivity of monocytes to cortisol is down-regulated by exer-cise (DeRijk et al. 1996). Dexamethasone inhibitionof lipopolysaccharide (LPS) -stimulated IL-6 pro-duction by monocytes is attenuated following anexercise bout (DeRijk et al. 1996; Smits et al. 1998).This attenuation allows monocytes to increase IL-6production, despite the anti-inflammatory influenceof cortisol. However, the production of severalproinflammatory cytokines by monocytes, includ-ing IL-6, is decreased and the production of the anti-inflammatory cytokines IL-10 and IFN-γ is notaffected by acute exercise (Smits et al. 1998). Thus,the influence of exercise on cytokine production iscomplex and further research is needed to clarifythis issue. It is important to remember that IL-6 synthesis by monocytes is less significant follow-ing strenuous exercise than that by skeletal muscle(Steensberg et al. 2002).

In response to exercise of light to moderate intensity or short duration, i.e. that in which cortisoldoes not increase and the effects of catecholaminesprevail, redistribution of immune cells is accompan-ied by a few modest shifts in immune cell function.B-cells are responsible for immunoglobulin (IgA,IgE, IgG and IgM) production and increased serumconcentrations of both IgA and IgG have been meas-ured after moderate exercise in some (Nehlsen-Cannarella et al. 1991) but not all studies (Eliakim et al. 1997). Prolonged exercise decreases the levelsof salivary IgA, an indication of decreased mucosalimmune defenses (Walsh et al. 2002). In the longterm, the process leading to development of anti-body titers upon vaccination is not influenced by a single stressful bout of exercise (Nieman 1997).Thus, moderate exercise in which the catechola-mines prevail do not appear to influence humoralimmunity to a significant extent, but prolongedstrenuous exercise in which the influence of cortisolprevails does decrease immune defense.

Cellular immunity may be lowered followingstrenuous exercise that includes the elevation of cor-tisol. Antigen presentation by macrophages appearsto be attenuated owing to suppressed expression of

Page 372: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

356 chapter 25

major histocompatibility complex II and this sup-pression correlates with cortisol elevations (Woodset al. 1997). Consistent with a reduction in the anti-gen presentation process is the measurement ofsmaller delayed-type hypersensitivity reactions following a half-marathon (Bruunsgaard et al. 1997b). This indicates that stressful exercise results in smaller cell-mediated immunity and antibodyproduction responses to antigenic challenge. Addi-tionally, glucocorticoids are capable of inducingapoptosis (programmed cell death) in lymphocytesand there is some evidence that strenuous exercisepromotes T-lymphocyte, lymphocyte, monocyteand neutrophil apoptosis in various compartmentsin both humans and rodents (Hsu et al. 2002; Moorenet al. 2002; Hoffman-Goetz & Quadrilatero 2003).Recent research in which carbohydrate and placebosupplementation elicited similar cortisol responsesimmediately post-exercise measured more apopto-sis in the placebo group (Green et al. 2003). This sug-gests that cortisol is not likely to be a causative factorfor apoptosis in response to exercise.

The effect of exercise on lymphocyte proliferationis unclear. The only conclusion that can be drawnregarding exercise-induced modulations in the ability of T- and B-lymphocytes to proliferate is that the data are mixed and inconsistent (Nielsen &Pedersen 1997). Similarly, the effects of strenuousexercise on T- and B-cell functions are mixed. How-ever, there are many reports of decreased prolifera-tion responses of T- and or B-lymphocytes to mitogenstimulation following strenuous, prolonged exer-cise (Green et al. 2003). Examination of the propor-tion of activated CD4+ and CD8+ T-lymphocytes inthe circulation expressing the CD69 surface marker,revealed that the proportion of activated lympho-cytes was consistent across pre- and post-exercisemeasurements (Green et al. 2003). Based on this, a decrease in T-cell proliferation would not beexpected, even following exercise that elicits anincrease in cortisol. Dohi et al. (2002) found that theproliferation response to a T- and B-cell mitogenwas lower in a group of women who had an increasein cortisol compared to those who did not have anincrease in cortisol, while there was no exercise-induced change in proliferation to T-cell mitogens.This suggests that B-cell proliferation may be a bit

more sensitive to cortisol and or strenuous exercise-induced inhibition.

The NKCA response to exercise must be con-sidered both when NK-cell concentrations areincreased, as during and immediately followingexercise, and when concentrations are decreased, as in the hours following strenuous or prolongedexercise. The mechanisms controlling the exercise-induced modulation of NKCA are likely to differduring each of these phases. For exercise of moder-ate intensity and duration, for example 45 min at50% of Vo2max, the increases in NK-cell concentra-tions and NKCA are of similar magnitude, suggest-ing that the increase in NKCA is the result ofincreased NK cells (Nieman et al. 1993b). At higherintensities, reports of the response of NKCA on aper NK cell basis are variable with some reportingan increase in per cell activity (Nieman et al. 1993b;Strasner et al. 1996), and others reporting a decreasein per cell activity (Nieman et al. 1995b; Nielsen et al.1996).

During the delayed phase of decreased NK-cell concentrations, NKCA may be reduced toapproximately half of pre-exercise activity for a fewhours. The decrease in NK-cell activity has beenattributed to decrease in NK-cell numbers (Nieman1997; Miles et al. 2002b), inhibition by prostaglandinE2 (Pedersen & Ullum 1994; Rhind et al. 1999) andinhibition by cortisol (Berk et al. 1990). However, the time frame for down-regulation of NKCA bycortisol does not match the time-course of exercise-induced changes in NKCA, thus cortisol is not con-sidered a likely modulator of NKCA beyond theeffects of circulating numbers (Nieman et al. 1993b,1995a).

One means of distilling out the influence of corti-sol is to examine the research comparing enduranceexercise with and without carbohydrate supple-mentation. Carbohydrate supplementation duringprolonged endurance exercise decreases the stresshormone response (Nieman et al. 1998, 2001; Greenet al. 2003). Thus, the difference in immune res-ponses between conditions may reflect the types offunctions influenced by cortisol, if the time course of differences in cortisol matches that of func-tional changes in immune cells. According to thisparadigm, it appears that cortisol increases may be

Page 373: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 357

associated with enhanced monocyte and neutrophiloxidative bursts (Nieman et al. 1998), phagocytosis(Henson et al. 2000), enhanced production of somecytokines, for example IL-8 and IL-10 (Nehlsen-Cannarella et al. 1997; Nieman et al. 2001), but notwith apoptosis (Green et al. 2003) or IL-6 (Nieman et al. 2001). Reports on the effect carbohydrate sup-plementation on lymphocyte proliferation are mixed(Henson et al. 1998; Mitchell et al. 1998). Given thetwo-way interaction between the immune systemand the HPA axis, it may be that the inflammatoryresponses drive the release of cortisol and not viceversa. Evidence to support this hypothesis is thatinfusion of recombinant IL-6 to match levels typ-ically measured during exercise elicits a substantialcortisol response (Steensburg et al. 2003).

Susceptibility to illness

The down-regulation of various immune defensesfollowing strenuous and prolonged exercise mayincrease the likelihood that a person will succumb toinfection. Athletes are more susceptible to infectiousillness, particularly upper respiratory tract infection(URTI), during periods of intense training and in the few weeks following particularly stressful com-petitive events such as a marathon (Peters &Bateman 1983; Nieman 1998). Reports of incidenceof URTI in athletes under these conditions vary, buta rough estimate may be that the risk is approx-imately double. That is, most athletes do not getURTI following strenuous competitions, but the riskis significantly increased (Nieman 2000). The down-time in training or the potential affect on perform-ance during URTI or other infections are issues ofsubstantial concern for many competitive athletes.Additionally, data from athletes may apply to otherphysically demanding situations, including occupa-tional or leisure time activities.

The mechanisms contributing to increased sus-ceptibility to illness have not been definitivelyidentified; however, a number of known responsesto strenuous exercise are suspected to contribute.The period following strenuous exercise in whichlymphocyte numbers and functions are decreased,nasal neutrophil phagocytosis is decreased, there isa decrease in salivary IgA and attenuation of anti-

gen presentation occurs and has been hypothesizedto be an ‘open window’ of opportunity for weakenedimmune defenses to be overcome by a virus (Nieman2000). The first line of defense against bacterial orviral pathogens, including mucosal immunoglobu-lins and the activity of neutrophils in the nasalmucosa, is weakened following strenuous exercise(Müns 1993). Similarly, salivary production of IgAis reduced following strenuous exercise (Mackinnonet al. 1989; Mackinnon & Jenkins 1993; Nieman et al. 2002), further adding to the impaired first line of defense against URTI. Decreased salivaryIgA is the only immune measure that has been asso-ciated with the onset of URTI (Mackinnon 2000).The tendency for both catecholamines and cortisolto inhibit type 1 immune responses and promotetype 2 responses tilts the scale toward increase susceptibility to URTI. This pattern of T-cell andcytokine responses has been definitively identifiedfollowing 2.5 h of treadmill running (Steensburg et al. 2001). Additionally, IL-6 produced duringexercise stimulates release of cortisol (Steensberg et al. 2003). Thus, there are a number of mechanismsby which the neuroendocrine and immune systemsare induced by strenuous and prolonged exercise tofavor Th2 and inflammatory responses at theexpense of the Th1 system. This shift decreases viraldefenses and may increase the ability of viruses toflourish, particularly those in the upper respiratorytract.

Exercise training

The relationship between exercise training or phys-ical activity and immune status depends on the dose of exercise and likely also on the opportunityfor recovery from the stress of exercise. There seemsto be general agreement that moderate levels oftraining may stimulate overall immune defenseswhile strenuous or severe training may suppressimmune defenses (Nieman 1997; Shephard 1997). Itis likely that the immune changes induced by train-ing are a component of a systemic response to stress,including the neuroendocrine response. Thus, theratio of stress to recovery may be a key componentdictating the shift from benefit to detriment as thestress of exercise training increases.

Page 374: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

358 chapter 25

Leukocyte trafficking

The number of leukocytes that reside in the circula-tion at rest may be used as a gross indication ofimmune status; however, the significance of modestfluctuations within or near the normal range has not been determined. Large deviations from thenormal range can be used to identify pathologies.For example, CD4+ T-cell concentrations can bemeasured to monitor the progression of humanimmunodeficiency virus/acquired immunodefici-ency syndrome (HIV/AIDS) (Hazenberg et al. 2003),and neutrophil concentrations can be measured to determine whether inflammation is occurring(Smith, L.L. & Miles 2000). Both longitudinal andcross-sectional studies have been employed todetermine whether exercise training significantlyimpacts concentrations of leukocyte subpopula-tions at rest. A number of studies report increasednumbers of neutrophils and NK cells in endurancetrained compared to non-trained individuals or inresponse to moderate exercise training (Nieman1997; Shephard 1997). Longitudinal studies of resist-ance training have not measured higher NK-cellconcentrations in response to training; however,one study measured an elevation after 3 months oftraining that did not persist through 6 months oftraining (Flynn et al. 1999; Miles et al. 2002a). Exer-cise training does not appear to have a consistentinfluence on resting concentrations of monocytes,CD4+ and CD8+ T-lymphocytes, or B-lymphocytes(Woods & Davis 1994; Shephard 1997; Miles et al.2002a). With training that is considered strenuous,typically by athletes at the national or internationallevels, decreased concentrations of monocytes havebeen reported (Shephard 1997).

The absolute magnitude of increases anddecreases in circulating leukocyte populations inresponse to exercise varies considerably amongstindividuals. The influence of exercise training onthe catecholamine response to exercise has not beendefinitively determined. Exercise training inducesan increase in work capacity that decreases the relat-ive proportion of maximal work intensity requiredfor any given absolute work effort. For example, theoxygen consumption associated with running at a 6-min per mile (1.609 km) pace may be 90% of Vo2max

before and 75% of Vo2max after endurance training.Thus, an equivalent absolute work rate requires alower relative proportion of the maximal effort aftertraining. This is true for virtually all types of train-ing. With respect to the neuroendocrine response toexercise, the rise in catecholamines and other hor-mones that occurred for this individual working atthe same absolute rate will be attenuated before andafter training (Kjær et al. 1988).

The influence of the neuroendocrine system onleukocyte trafficking during exercise is a function of the relative intensity and duration of the exercise.It is the relative exercise intensity rather than theabsolute exercise intensity that dictates the neuro-endocrine and consequent immune responses toexercise. Research data consistent with this effect of exercise training on the immune response toacute exercise have been presented for enduranceexercise, resistance exercise and high-intensityinterval exercise (Moyna et al. 1996; Shephard 1997;Miles et al. 2002a).

Cytokines

Resting plasma cytokine concentrations typically do not differ between trained and untrained indi-viduals (Smith, J.A. et al. 1992; Shephard 1997).However, higher IL-1β and IL-6 concentrationshave been reported in endurance trained comparedto untrained individuals in some studies (Sprengeret al. 1992; Mucci et al. 2000). This may be a functionof decreased sensitivity to cortisol inhibition in thetrained individuals (Duclos et al. 1999) or perhapsthe cumulative effects of exercise-induced inflam-mation (Shephard 1997).

Leukocyte function

While basal levels of the HPA axis hormones ACTHand cortisol do not appear to be influenced byendurance training, decreased sensitivity of mono-cytes to cortisol has been measured in endurancetrained compared to untrained men (Duclos et al.1999, 2001). Production of IL-6 by monocytes stimu-lated with LPS is greater for endurance trained compared to untrained men (Duclos et al. 1999).However, following an acute bout of endurance

Page 375: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 359

exercise, the sensitivity of monocytes to glucocor-ticoids for endurance trained men increased toapproximately that of the untrained men (Duclos et al. 1999). Thus, the sensitivity of monocytes to cor-tisol is enhanced by endurance training, but is not astatic trait. The ramifications of this may reach a vastarray of immune functions, as monocytes produce anumber of cytokines that potentially impact allimmune cells. In contrast, 12 weeks of progressiveresistance training did not influence the stimulatedproduction of TNF-α, IL-1β, IL-2 or IL-6 by peri-pheral blood mononuclear cells (PBMCs) collectedfrom younger and older healthy subjects or subjectswith rheumatoid arthritis (Rall et al. 1996). Thus, theability of leukocytes to produce cytokines does notappear to be substantially impacted by exercisetraining, with the possible exception of monocytes.

A differential response to exercise has been iden-tified between trained and untrained individualswith respect to neutrophil adherence and oxidativeburst (Pyne 1994; Shephard 1997). While this is con-sistent with increased sensitivity to cortisol, thisaspect of the neutrophil response to exercise train-ing has not been investigated. There is no consist-ency amongst reports of other aspects of neutrophilfunction in response to exercise training or in cross-sectional comparisons of trained and untrainedindividuals (Shephard 1997).

Functional activity of T- and B-lymphocytes is notsignificantly altered in response to exercise trainingor in cross-sectional comparisons of trained anduntrained subjects (Shephard 1997). Longitudinalstudies indicate that this is consistent for bothyounger and older individuals and for enduranceand resistance training (Rall et al. 1996; Woods et al.1999; Miles et al. 2002a), although Woods et al. (1999)measured a modest enhancement of T-lymphocyteproliferation following 6 months of aerobic trainingin an elderly population. With respect to B-cell pro-duction of immunoglobulins, levels in serum andsaliva are comparable in trained versus untrainedindividuals across the vast majority of comparisons,both cross-sectional and longitudinal (Shephard1997; Potteiger et al. 2001). One exception to this isthat decreased salivary IgA has been measured inathletes undergoing particularly intensive portionsof their training, i.e. when training volume and

intensity are greatest, in many, but not all, investiga-tions of this phenomenon (Tharp & Barnes 1990;Mackinnon & Hooper 1994; Gleeson et al. 1999; Pyneet al. 2000). The endocrine system may be partlyresponsible for this shift, that also may includeshifts in cell numbers and reactivity (B-, CD4+ andCD8+ T-cells), the response to tissue damage andoverall stress (Shephard 1997).

A number of investigations report that exercisetraining increases the activity level of individual NK cells such that greater NKCA per NK cell in thecirculation is consistently measured (Pedersen et al.1989; Nieman et al. 1990, 1993a; Woods et al. 1999).Recent research using a rodent model (spontan-eously hypertensive rats) suggests that: (i) there is adose–response in which in vivo NKCA is enhancedas exercise volume increases, but this plateaus offand no further gains are achieved beyond moderatevolumes; and (ii) the gains in NKCA can be abol-ished if sufficient rest and recovery do not occur( Jonsdottir & Hoffmann 2000). This information, inconjunction with the research relating to the acuteeffects of strenuous exercise, suggests that super-imposing new exercise stresses before completerecovery has occurred may lead to an apparently‘chronic’ suppression. It is difficult to separate out adaptations to exercise training over time fromlingering effects of the most recent exercise sessionfor athletes undergoing strenuous training (Shephard1997).

Overtraining is a situation in which training volume or intensity exceeds the capacity of the bodyto adapt and a feeling of fatigue and performancedecrements are typical (Urhausen et al. 1995; Smith,L.L. 2000). Common characteristics associated withovertraining are sympathetic nervous system andHPA axis imbalances and an increased susceptib-ility to illnesses (Fitzgerald 1991; Urhausen et al.1995). This is not surprising given the interrelated-ness of these systems. Smith, L.L. (2000) hypothes-ized that tissue trauma, particularly muscle damage,may result in increased inflammatory cytokines, fol-lowed by activation of the HPA axis, which resultsdown-regulation of immune cell function. Further,she suggests that this represents an advanced stageof the general adaptation model to stress proposedby Selye, and may be the point at which recovery

Page 376: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

360 chapter 25

and survival are more important than adaptation to stress. This model is consistent with the data pre-sented in this chapter; however, more recent researchto test this hypothesis did not measure increasedlevels of inflammatory cytokines in athletes duringa period of induced overtraining (Halson et al. 2003).The complexities of the neuroendocrine and immunesystems make it very difficult to verify or reject sev-eral components of this hypothesis.

Susceptibility to illness

The relationship between levels of exercise trainingand physical activity to incidence of illness is con-sistent with the overall pattern of changes identifiedin immune parameters. That is, moderate trainingmay reduce susceptibility to illness somewhat, orreduce the duration and severity of symptoms(Nieman et al. 1990). Recent epidemiological studiesalso confirmed a similar relationship between phys-ical activity and URTI in a diverse, healthy, adultpopulation (Matthews et al. 2002) and school chil-dren in Poland (Jedrychowski et al. 2001). Consist-ent with this trend toward lower URTI incidencewith moderate levels of physical activity or exercisetraining is the measurement of increased salivaryIgA in response to moderate exercise training(Klentrou et al. 2002).

When training intensifies and reaches high volumes and or intensities, perhaps coupled withlimited recovery, then there is an increase in suscept-ibility to URTI (Shephard 1997; Mackinnon 2000).Recent research with elite Australian swimmers hasadvanced IgA levels and stress-induced viral react-ivation of dormant viruses, such as Epstein–Barrvirus, as likely mechanisms contributing to the incidence of URTI during high level training(Gleeson et al. 2002). In a study of training volumeand tennis players, salivary IgA levels were inverselyrelated to training volume, but IgA levels were notpredictive of URTI incidence. Researchers continueto search for the link between training levels andimmunosuppression. Illness occurrence is likely tobe a multidimensional function of the temporalaccumulation of acute, exercise-induced immuno-suppression from multiple training sessions, thelevel of recovery from bout to bout, nutritional

factors, additional stressors and exposure to virusand/or the reactivation of dormant viruses. As notedpreviously, the ‘the general adaptation syndrome’hypothesis of Selye (1936) proposes that all stressorselicit some common responses, including activationof the HPA axis. Thus, exercise may best be viewedas one component of the total stress to which a per-son must respond.

Cytokine control of muscle responses tostress and damage

Muscle cells release cytokines in response to exer-cise, whether or not cellular damage occurs. Currentresearch suggests that many of these cytokines haveregulatory functions and may be necessary signalsfor various cellular functions and as systemic signals to the brain, particularly to the HPA axis.That is, inflammation is just one of the functions inwhich these molecules, particularly IL-6 and LIF,participate, and the production of these moleculeswithin muscle is not dependent on tissue injury. Forexample, LIF has a hypertrophic effect on skeletalmuscle cells and induces satellite cell prolifera-tion (Spangenburg & Booth 2002; Gregorevic et al.2002). IL-6 may act as a systemic signal reflectinglow glycogen levels in exercising muscle cells(MacDonald et al. 2003). However, when muscledamage does occur, IL-6 and LIF participate in theinflammatory response. As indicated previously,TNF-α, IL-1β, IL-6 and LIF are capable of stimulat-ing the HPA axis in a negative feedback loop toinduce production of the anti-inflammatory hor-mone, cortisol (Mastorakos et al. 1993; Chesnokova& Melmed 2002; Tsigos & Chrousos 2002). Unfortu-nately, the response of LIF to exercise scarcely hasbeen investigated and little is known at this point.

The type of muscle damage most typically associ-ated with exercise is characterized by delayed onsetmuscle soreness (DOMS); for example, the sorenessfelt in muscles the day following the performance of exercise to which one is not accustomed. Indir-ect indicators of this damage are DOMS, loss ofstrength, swelling and elevations in serum creatinekinase activity (Miles & Clarkson 1994). The exercise-induced inflammatory response involving tissuedamage likely occurs in the following sequence:

Page 377: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 361

(i) tissue injury/stress; (ii) resident macrophageactivation; (iii) release of inflammatory ‘alarm’cytokines TNF-α and IL-1β; (iv) stimulation of localrelease of chemoattractants, for example IL-8; (v)local production of IL-6; (vi) stimulation of acutephase proteins by the liver; (vii) stimulation of theHPA axis; and (viii) leukocytosis and leukocytemigration to site of injury (Smith, L.L. & Miles 2000).IL-1β and TNF-α initiate the inflammatory cascade,which includes production of IL-6, which in turnstimulates production of a number of counter-inflammatory cytokines and other molecules,including IL-10 and IL-1ra (Turnbull et al. 1994;Smith, L.L. & Miles 2000). Local production of IL-1βhas been demonstrated in muscle biopsy samplesfollowing eccentric exercise associated with muscledamage (Malm et al. 2000); however, the increases in blood are short-lived and typically small relativeto other cytokines such as IL-6 or IL-10 (Shephard1997; Suzuki et al. 2002). Both IL-1β and IL-6 act asgrowth factors to promote regeneration at the site oftissue damage (Northoff et al. 1995).

Within the realm of inflammation, the process of regeneration is dependent on the removal ofdamaged cellular debris by phagocytes. The cellu-lar response to exercise-induced muscle damageincludes infiltration by and activation of localmacrophages (Stupka et al. 2001; LaPointe et al.2002). Infiltration of injured tissue by neutrophilshas been measured in some (Brickson et al. 2001;MacIntyre et al. 2001), but not all investigations(LaPointe et al. 2002). Muscle biopsy data suggeststhat this process occurs over a period of weeks(Lieber 1992). This certainly exceeds the duration ofany detectable markers of inflammation in the sys-temic circulation. Thus, much of the response toexercise-induced muscle injury occurs locally ratherthan systemically.

If exercise is prolonged and strenuous or mechan-ically stressful, for example containing a high-forceeccentric component, then some degree of tissuedamage may be expected and the components of thedescribed response are measurable. Marathons andcomparable events induce skeletal muscle damageevidenced by disruption of myofibrillar organiza-tion and an efflux of the intramuscular enzyme cre-atine kinase from muscle to blood (Rogers et al. 1985;

Warhol et al. 1985). Following marathon-type races,increases in plasma or serum TNF-α, IL-1β, IL-6, IL-10 and IL-1ra have been measured (Northoff et al. 1994; Drenth et al. 1995; Nehlsen-Cannarella et al. 1997; Ostrowski et al. 1998a, 1998b; Henson et al.2000; Nieman et al. 2001). High force eccentric exer-cise also induces skeletal muscle damage (Fridén & Lieber 2001) and smaller increases in IL-1β, IL-6and IL-1ra (Bruunsgaard et al. 1997a; Smith, L.L. et al.2000; Chen & Hsieh 2001; Toft et al. 2002). Increasesin TNF-α typically are not measured (Bruunsgaardet al. 1997a; Toft et al. 2002), but this may reflect thedifficulty of measuring a systemic response to alocal inflammatory response that is relatively smallin magnitude. The prolonged period of inflamma-tion within muscles is not accompanied by eleva-tions in cortisol (Pizza et al. 1995; Lenn et al. 2002).Thus, under reasonable circumstances, the inflam-matory response to muscle damage does not appearsubstantial enough to stimulate the HPA axis for anextended period of time. However, there is a neuro-endocrine and immune basis for the cytokineresponse to muscle damage to stimulate the HPAaxis if IL-6, and possibly TNF-α, IL-1β or LIF, pro-duction was substantial.

The hypothalamus uses the neuroendocrine–immune network to integrate exercise-induced stresssignals and central signals to induce appropriateresponses to those stresses (Steinacker et al. 2004).Recent muscle biopsy data suggest a link betweenincreases in circulating cortisol and activation ofproteolysis within the injured muscle (Willoughbyet al. 2003). Exercise-induced injury is mediated bythe ubiquitin-proteolytic pathway, and cortisol up-regulates gene expression of several components ofthis pathway, for example ubiquitin itself, ubiquitinconjugating enzyme and glucocorticoid receptors.Consistent with the protective effects known to occurafter a single exercise bout is the down-regulation of this system following a second bout of the sameinjurious exercise. Thus, the two-way network ofcommunication involves cytokine signals generatedlocally to signal the HPA axis and HPA axis signalsgenerated centrally to signal the muscle to repairand regenerate in response to tissue injury.

The role of inflammatory mediators in the res-ponse to exercise-induced muscle damage has been

Page 378: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

362 chapter 25

called into question in light of research in whichanti-inflammatory treatment modalities failed tohave a substantial impact on DOMS or the recoveryof function (Cheung et al. 2003). However, theresearch cited above is a small sample of the evid-ence of various components of the inflammatoryresponse measured in response to exercise-inducedmuscle damage. Thus, it may be prudent simply tosuggest that the influence of prostaglandin E2, thetypical target of anti-inflammatory medications, isnot a major player in the inflammatory response(Semark et al. 1999). As a result, anti-inflammatorytreatments do not appear to interfere with orenhance the healing process and have only a minorimpact on soreness (Cheung et al. 2003).

Summary

The immune and neuroendocrine responses to thestress of exercise certainly are linked. However,modulation of exercise-induced immune responsesalso is influenced by the inflammatory response to tissue injury. Cytokines mediating the local andsystemic responses to exercise are often multifunc-tional, for example IL-6. Many of the functions ofthese cytokines are unrelated to inflammation or

immune modulation per se, for example LIF may beacting as a growth factor and have nothing to dowith tissue injury. The two-way communicationnetwork between the neuroendocrine and immunesystems is such that cytokines produced by musclesare capable of activating the HPA axis. Reciprocalregulation of the neuroendocrine and immune sys-tems is infinitely complex. Regardless, research stud-ies are consistent in measuring enhanced immunefunction in response to moderately stressful acuteexercise or exercise training. The level of exercisestress that the body can tolerate without immuno-suppression and increased susceptibility to illness islikely to vary according to the level of other stressesthat the body must endure. A number of studieshave measured suppressed function in some com-ponents of immunity and increased URTI occur-rences in athletes when the physical demands oftraining and competition are extreme. In light of the interactions between the neuroendocrine andimmune systems, individuals wishing to maximizetheir training volumes and intensities may be welladvised to reduce the stress response to exercise viaadequate recovery and various nutritional strateg-ies and to minimize other forms of stress to whichthe body must respond.

ReferencesAshwell, J.D., Lu, F.W.M. & Vacchio, M.S.

(2000) Glucocorticoids in T-celldevelopment and function. AnnualReview of Immunology 18, 309–345.

Atalay, M., Marnila, P., Lilius, E.-M.,Hanninen, O. & Sen, C.K. (1996)Glutathione-dependent modulation ofexhausting exercise-induced changes in neutrophil function in rats. EuropeanJournal of Applied Physiology 74, 342–347.

Beishuizen, A. & Thijs, L.G. (2003)Endotoxin and the hypothalamo–pituitary–adrenal (HPA) axis. Journal ofEndotoxin Research 9, 3–24.

Benschop, R.J., Nieuwenhuis, E.E.S.,Tromp, E.A.M. et al. (1994) Effects of β-adrenergic blockade on immunologicand cardiovascular changes induced bymental stress. Circulation 89, 762–769.

Berk, L.S., Nieman, D.C., Youngberg, W.S.et al. (1990) The effect of long endurancerunning on natural killer cells inmarathoners. Medicine and Science inSports and Exercise 22, 207–212.

Blank, S.E., Jones, T.B., Lee, E.G. et al.(1997) Modulation of NK cell cytolyticactivity by macrophages in chronicallyexercise-stressed mice. Journal of AppliedPhysiology 83, 845–850.

Borger, P., Hoekstra, Y., Esselink, M.T. et al. (1998) Beta-adrenoceptor-mediatedinhibition of IFN-γ, IL-3, and GM-CSFmRNA accumulation in activatedhuman T lymphocytes is solelymediated by the β2-adrenoceptorsubtype. American Journal of Respiratory,Cellular, and Molecular Biology 19,400–407.

Boxer, L.A., Allen, J.M. & Baehner, R.L.(1980) Diminished polymorphonuclearleukocyte adherence. Journal of ClinicalInvestigation 66, 268–274.

Brenner, I., Shek, P.N., Zamecnik, J. &Shephard, R.J. (1998) Stress hormonesand the immunological response to heatand exercise. International Journal ofSports Medicine 19, 130–143.

Brickson, S. Hollander, J. Corr, D.T., Ji, L.L.

& Best, T.M. (2001) Oxidant productionand immune response after stretchinjury in skeletal muscle. Medicine andScience in Sports and Exercise 33,2010–2015.

Bruunsgaard, H., Galbo, H., Halkjaer-Kristensen, J. et al. (1997a). Exercise-induced increase in interleukin-6 isrelated to muscle damage. Journal ofPhysiology 499, 833–841.

Bruunsgaard, H., Hartkopp, A., Mohr, T. et al. (1997b) In vivo cell-mediatedimmunity and vaccination responsefollowing prolonged, intense exercise.Medicine and Science in Sports and Exercise29, 1176–1181.

Bruunsgaard, H., Jensen, M.S., Schjerling,P. et al. (1999) Exercise inducesrecruitment of lymphocytes with anactivated phenotype and short telomeresin young and elderly humans. LifeSciences 65, 2623–2633.

Buckingham, J.C., Loxley, H.D., Christian,H.C. & Philip, J.G. (1996) Activation of

Page 379: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 363

the HPA axis by immune insults: rolesand interactions of the cytokines,eicosanoids, and glucocorticoids.Pharmacology Biochemistry and Behavior54, 285–298.

Cancedda, C., Filaci, G., Puppo, F. et al.(2002) Immune homeostasis requiresseveral biologic factors includingglucocorticoid hormones. Annals of the New York Academy of Sciences 966,49–63.

Carr, D.J.J. & Blalock, J.E. (1990)Neuropeptide hormones and receptorscommon to the immune andneuroendocrine systems: bidirectionalpathway of intersystem communication.In: Psychoneuroimmunology, 2nd edn.Academic Press, New York: 573–588.

Chen, T.C. & Hsieh, S.S. (2001) Effects of a 7-day eccentric training period on muscle damage and inflammation.Medicine and Science in Sports and Exercise33, 1732–1738.

Chesnokova, V. & Melmed, S. (2000)Leukemia inhibitory factor mediates the hypothalamic pituitary adrenal axisresponse to inflammation. Endocrinology141, 4032–4040.

Chesnokova, V. & Melmed, S. (2002)Minireview: Neuro-immuno-endocrinemodulation of the hypothalamic–pituitary–adrenal (HPA) axis by gp130signaling molecules. Endocrinology 143,1571–1574.

Chesnokova, V., Kariagina, A. & Melmed,S. (2002) Opposing effects of pituitaryleukemia inhibitory factor and SOCS-3on the ACTH axis response toinflammation. American Journal ofPhysiology, Endocrinology and Metabolism282, E111–E118.

Cheung, K., Hume, P.A. & Maxwell, L.(2003) Delayed onset muscle soreness:Treatment strategies and performancefactors. Sports Medicine 33, 145–164.

DeRijk, R.H., Petrides, J., Deuster, P., Gold, P.W. & Sternberg, E.M. (1996)Changes in corticosteroid sensitivity of peripheral blood lymphocytes afterstrenuous exercise in humans. Journal ofClinical Endocrinology and Metabolism 81,228–235.

Deuster, P.A., Zelazowska, E.B., Singh, A.,& Sternberg, E.M. (1999) Expression oflymphocyte subsets after exercise anddexamethasone in high and low stressresponders. Medicine and Science in Sports and Exercise 31, 1799–1806.

Dohi, K., Mastro, A.M., Miles, M.P. et al.(2002) Lymphocyte proliferation inresponse to acute heavy resistanceexercise in women: influence of muscle

Gleeson, M., Pyne, D.B., Austin, J.P. et al.(2002) Epstein–Barr virus reactivationand upper-respiratory illness in eliteswimmers. Medicine and Science in Sportsand Exercise 34, 411–417.

Green, K.J., Croaker, S.J. & Rowbottom,D.G. (2003) Carbohydratesupplementation and exercise-inducedchanges in T-lymphocyte function.Journal of Applied Physiology 95,1216–1223.

Gregorevic, P., Williams, D.A. & Lynch,G.S. (2002) Effects of leukemia inhibitoryfactor on rat skeletal muscles aremodulated by clenbuterol. Muscle andNerve 25, 194–201.

Haddad, J.J., Saade, N.E. & Safieh-Garabedian, B. (2002) Cytokines andneuro-immune-endocrine interactions: a role for the hypothalamic–pituitary–adrenal revolving axis. Journal ofNeuroimmunology 133, 1–19.

Halson, S.L., Lancaster, G.I., Jeukendrup,A.E. & Gleeson, M. (2003)Immunological responses tooverreaching in cyclists. Medicine andScience in Sports and Exercise 35, 854–861.

Hazenberg, M.D., Otto, S.A., van Benthem,B.H. et al. (2003) Persistent immuneactivation inf HIV-1 infection isassociated with progression of AIDS.AIDS (London, England) 17, 1881–1888.

Henson, D.A., Nieman, D.C., Parker, J.C.D. et al. (1998) Carbohydratesupplementation and the lymphocyteproliferative response to long endurancerunning. International Journal of SportsMedicine 19, 574–580.

Henson, D.A., Nieman, D.C., Nehlsen-Cannarella, S.L. et al. (2000) Influence of carbohydrate on cytokine andphagocytic response to 2 h of rowing.Medicine and Science in Sports and Exercise32, 1384–1389.

Hetherington, S.V. & Quie, P.G. (1985)Human polymorphonuclear leukocytesof the bone marrow, circulation, andmarginated pool: function and granuleprotein content. American Journal ofHematology 20, 235–246.

Hoffman-Goetz, L. & Quadrilatero, J.(2003) Treadmill exercise in miceincreases intestinal lymphocyte loss viaapoptosis. Acta Physiologica Scandanavica179, 289–297.

Hsu, T.-G., Hsu, K.-M., Kong, C.-W. et al. (2002) Leukocyte mitochondriaalterations after aerobic exercise intrained human subjects. Medicine andScience in Sports and Exercise 34, 438–442.

Ibfelt, T., Petersen, E.W., Bruunsgaard, H.,Sandmand, M. & Pedersen, B.K. (2002)

strength and total work. European Journalof Applied Physiology 85, 367–373.

Drenth, J.P.H., Van Uum, S.H.M., Van Deuren, M. et al. J.W.M. (1995)Endurance run increases circulating IL-6and IL-1ra but downregulates ex vivoTNF-α and IL-1β production. Journal of Applied Physiology 79, 1497–1503.

Duclos, M., Minkhar, M., Sarrieau, A. et al. (1999) Reversibility of endurancetraining-induced changes onglucocorticoid sensitivity of monocytesby an acute exercise. ClinicalEndocrinology 51, 749–756.

Duclos, M., Corcuff, J.-B., Pehourcq, F. &Tabarin, A. (2001) Decreased pituitarysensitivity to glucocorticoids inendurance-trained men. EuropeanJournal of Endocrinology 144, 363–368.

Elenkov, I.J., Wilder, R.L., Chrousos, G.P.& Vizi, E.S. (2000) The sympatheticnerve: an integrative interface betweentwo supersystems: the brain and theimmune system. Pharmacological Reviews52, 595–638.

Eliakim, A., Wolach, B. Kodesh, E. et al.(1997) Cellular and humoral immuneresponse to exercise among gymnastsand untrained girls. International Journalof Sports Medicine 18, 208–212.

Febbraio, M.A., Ott, P., Nielsen, H.B. et al. (2003a) Hepatosplanchnic clearanceof interleukin-6 in humans duringexercise. American Journal of Physiology,Endocrinology and Metabolism 285,E397–E402.

Febbraio, M.A., Steensburg, A., Starkie,R.L., McConell, G.K. & Kingwell, B.A.(2003b). Skeletal muscle interleukin-6and tumor necrosis factor-α release inhealthy subjects and patients with type 2diabetes at rest and during exercise.Metabolism 52, 939–944.

Fitzgerald, L. (1991) Overtraining increases the susceptibility to infection.International Journal of Sports Medicine 12,S5–S8.

Flynn, M.G., Fahlman, M., Braun, W.A. et al. (1999) Effects of resistance trainingon selected indexes of immune functionin elderly women. Journal of AppliedPhysiology 86, 1905–1913.

Fridén, J. & Lieber, R.L. (2001) Eccentricexercise-induced injuries to contractileand cytoskeletal muscle fibrecomponents. Acta PhysiologicaScandanavica 171, 321–326.

Gleeson, M., McDonald, W.A., Pyne, D.B.et al. (1999) Salivary IgA levels andinfection risk in elite swimmers.Medicine and Science in Sports and Exercise31, 67–73.

Page 380: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

364 chapter 25

Exercise-induced change in type 1cytokine-producing CD8+ T cells inrelated to a decrease in memory T cells. Journal of Applied Physiology 93,645–648.

Jedrychowski, W., Maugeri, U., Flak, E.,Mroz, E. & Bianchi, I. (2001) Cohortstudy on low physical activity level andrecurrent acute respiratory infections inschoolchildren. Central European Journalof Public Health 9, 126–129.

Jonsdottir, I.H. & Hoffmann, P. (2000) Thesignificance of intensity and duration of exercise on natural immunity in rats.Medicine and Science in Sports and Exercise32, 1908–1912.

Jonsdottir, I.H. Schjerling, P., Ostrowski, K.et al. (2000) Muscle contractions induceinterleukin-6 mRNA production in ratskeletal muscles. Journal of Physiology528, 157–163.

Kappel, M., Tvede, N., Galbo, H. et al.(1991) Evidence the the effect of physicalexercise on NK cell activity is mediatedby epinephrine. Journal of AppliedPhysiology 70, 2530–2534.

Kappel, M., Poulsen, T.D., Galbo, H. &Pedersen, B.K. (1998) Influence of minorincreases in plasma catecholamines onnatural killer cell activity. HormoneResearch 49, 22–26.

Kjær, M. & Dela, F. (1996) Endocrineresponses to exercise. In: Exercise andImmune Function (Hoffman-Goetz, L.,ed.). CRC Press, Boca Raton, FL: 1–20.

Kjær, M., Bangsbo, J., Lortie, G. & Galbo,H. (1988) Hormonal responses toexercise in humans: influence of hypoxia and physical training. AmericanJournal of Physiology 254, R197–R203.

Klentrou, P., Cieslak, T., MacNeil, M.,Vintinner, A. & Plyley, M. (2002) Effectof moderate exercise on salivaryimmunoglobulin A and infection risk in humans. European Journal of AppliedPhysiology 87, 153–158.

Klokker, M., Kjær, M., Secher, N.H. et al.(1995) Natural killer cell response toexercise in humans: effect of hypoxiaand epidural anesthesia. Journal ofApplied Physiology 78, 709–716.

Klokker, M., Secher, N.H., Madsen, P.,Pedersen, M. & Perdersen, B.K. (1997)Adrenergic β1 and β1+2-receptorblockade suppress the natural killer cell response to head-up tilt in humans.Journal of Applied Physiology 83,1492–1498.

Kohm, A.P. & Sanders, V.M. (2001)Norepinephrine and β2-adrenergicreceptor stimulation regulate CD4+ Tand B lymphocyte function in vitro and

Mackinnon, L.T. & Jenkins, D.G. (1993)Decreased salivary immunoglobulinsafter intense interval exercise before andafter training. Medicine and Science inSports and Exercise 25, 678–683.

Mackinnon, L.T., Chick, T.W., van As, A. &Tomasi, T.B. (1989) Decreased secretoryimmunoglobulins following intenseendurance exercise. Sports Training,Medicine and Rehabilitation 1, 209–218.

McMurray, R.G. & Hackney, A.C. (2000)Endocrine responses to exercise andtraining. In: Exercise and Sport Science(Garrett, W.E. & Kirkendall, D.T., eds.).Lippincott, Williams & Wilkins,Philadelphia, PA: 135–161.

Maisel, A.S., Harris, C., Rearden, D.A. &Michel, M.C. (1990) Beta-adrenergicreceptors in lymphocyte subsets after exercise. Alterations in normalindividuals and patients with congestiveheart failure. Circulation 82, 2003–2010.

Malarkey, W.B., Wang, J., Cheney, C.,Glaser, R. & Nagaraja, H. (2002). Humanlymphocyte growth hormone stimulatesinterferon γ production and is inhibitedby cortisol and epinephrine. Journal ofNeuroimmunology 123, 180–187.

Malm, C., Nyberg, P., Engström, M. et al.(2000) Immunological changes in humanskeletal muscle and blood after eccentricexercise and multiple biopsies. Journal ofPhysiology 529, 243–262.

Masek, K., Slánsky, J. Petrovicky, P. &Hadden, J.W. (2003) Neuroendocrineimmune interactions in health and disease. InternationalImmunopharmacology 3, 1235–1246.

Mastorakos, G., Chrousos, G.P. & Weber,J.S. (1993) Recombinant interleukin-6activated the hypothalamic–pituitary–adrenal axis in humans. Journal ofClinical Endocrinology and Metabolism 77,1690–1694.

Matthews, C.E., Ockene, I.S., Freedson,P.S. et al. (2002) Moderate to vigorousphysical activity and risk of upper-respiratory tract infection. Medicine and Science in Sports and Exercise 34,1242–1248.

Miles, M.P. & Clarkson, P.M. (1994)Exercise-induced muscle pain, soreness,and cramps. Journal of Sports Medicineand Physical Fitness 34, 203–216.

Miles, M.P., Leach, S.K., Kraemer, W.J. et al. (1998) Leukocyte adhesionmolecule expression during intenseresistance exercise. Journal of AppliedPhysiology 84, 1604–1609.

Miles, M.P., Kraemer, W.J., Grove, D.S. et al. (2002a) Effects of resistance trainingon resting immune parameters in

in vivo. Pharmacological Reviews 53,487–525.

LaPointe, B.M., Frenette, J. & Côté, C.(2002) Lengthening contraction-inducedinflammation is linked to secondarydamage but devoid of neutrophilinvasion. Journal of Applied Physiology92, 1995–2004.

Lenn, J., Uhl, T., Mattacola, C. et al. (2002)The effects of fish oil and isoflavones ondelayed onset muscle soreness. Medicineand Science in Sports and Exercise 34,1605–1613.

Leonard, B.E. & Song, C. (1996) Stress andthe immune system in the etiology ofanxiety and depression. PharmacologyBiochemistry and Behavior 54, 299–303.

Levine, S.J., Benfield, T. & Shelhamer, J.H.(1996) Corticosteroids induceintracellular interleukin-1 receptorantagonist type I expression by a humanairway epithelial cell line. AmericanJournal of Respiratory Molecular andCellular Biology 15, 245–251.

Lieber, R.L. (1992) Skeletal Muscle Structureand Function. Williams & Wilkins,Baltimore, MD.

Liles, W.C. & Van Voorhis, W.C. (1995)Review: nomenclature and biologicsignificance of cytokines involved in inflammation and host immuneresponse. Journal of Infectious Diseases172, 1573–1580.

MacDonald, C., Wojtaszewski, J.F.P.,Pedersen, B.D., Kiens, B. & Richter, E.A.(2003) Interleukin-6 release from humanskeletal muscle during exercise: relationto AMPK activity. Journal of AppliedPhysiology 95, 2273–2277.

McFarlin, B.K., Mitchell, J.B., McFarlin,M.A. & Steinhoff, G.M. (2003) Repeatedendurance exercise affects leukocytenumber but not NK cell activity.Medicine and Science in Sports and Exercise 35, 1130–1138.

MacIntyre, D.L., Sorichter, S., Mair, J.,Berg, A. & McKenzie, D.C. (2001)Markers of inflammation andmyofibrillar proteins following eccentric exercise in humans. EuropeanJournal of Applied Physiology 84, 180–186.

Mackinnon, L.T. (2000) Chronic exercisetraining effects on immune function.Medicine and Science in Sports and Exercise 32, S369–S376.

Mackinnon, L.T. & Hooper, S.L. (1994)Mucosal (secretory) immune systemresponses to exercise of varyingintensity and during overtraining.International Journal of Sports Medicine 15,S179–S183.

Page 381: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 365

women. European Journal of AppliedPhysiology 87, 506–508.

Miles, M.P., Mackinnon, L.T., Grove, D.S.et al. (2002b) The relationship of naturalkiller cell counts, perforin mRNA, andCD2 expression to post-exercise naturalkiller cell activity in humans. ActaPhysiologica Scandanavica 174, 317–325.

Mitchell, J.B., Pizza, F.X., Paquet, A. et al.(1998) Influence of carbohydrate statuson immune responses before and afterendurance exercise. Journal of AppliedPhysiology 84, 1917–1925.

Mitchell, J.B., Dugas, J.P., McFarlin, B.K. & Nelson, M.J. (2002) Effect of exercise,heat stress, and hydration on immunecell number and function. Medicine and Science in Sports and Exercise 34,1941–1950.

Mooren, F.C., Blöming, D., Lechtermann,A., Lerch, M.M. & Völker, K. (2002)Lymphocyte apoptosis after exhaustiveand moderate exercise. Journal of AppliedPhysiology 93, 147–153.

Mosmann, T.R. & Sad, S. (1996) Theexpanding universe of T-cell subsets:Th1, Th2 and more. Immunology Today17, 138–146.

Moyna, N.M., Acker, G.R., Weber, K.M. et al. (1996). The effects of incrementalsubmaximal exercise on circulatingleukocytes in physically active andsedentary males and females. EuropeanJournal of Applied Physiology 74, 211–218.

Mucci, P., Durand, F., Level, B., Bousquet,J. & Préfaut, C. (2000). Interleukins 1-β, -8, and histamine increases in highlytrained, exercising athletes. Medicine and Science in Sports and Exercise 32,1094–1100.

Müns, G. (1993) Effect of long-distancerunning on polymorphonuclearneutrophil phagocytic function of theupper airways. International Journal ofSports Medicine 15, 96–99.

Nehlsen-Cannarella, S.L., Nieman, D.C.,Jessen, J. et al. (1991) The effects of acutemoderate exercise on lymphocytefunction and serum immunoglobulinlevels. International Journal of SportsMedicine 12, 391–398.

Nehlsen-Cannarella, S.L., Fagoaga, O.R.,Nieman, D.C. et al. (1997) Carbohydrateand the cytokine response to 2.5 h ofrunning. Journal of Applied Physiology 82,1662–1667.

Nielsen, H.B. & Pedersen, B.K. (1997)Lymphcyte proliferation in response to exercise. European Journal of AppliedPhysiology 75, 375–379.

Nielsen, H.B., Secher, N.H., Kappel, M.,Hanel, B. & Pedersen, B.K. (1996)

Northoff, H., Weinstock, C. & Berg, A.(1994) The cytokine response tostrenuous exercise. International Journalof Sports Medicine 15, S167–S171.

Northoff, H., Enkel, S. & Weinstock, C.(1995) Exercise, injury and immunefunction. Exercise Immunology Review1, 1–25.

Ortega, E., Rodrigues, M.J., Barriga, C. & Forner, M.A. (1996) Corticosterone,prolactin and thyroid hormone ashormonal mediators of the stimulatedphagocytic capacity of peritonealmacrophages after high-intensityexercise. International Journal of SportsMedicine 17, 149–155.

Ortega, E., Forner, M.A. & Barriga, C.(1997) Exercise-induced stimulation of murine macrophage chemotaxis: role of corticosterone and prolactin asmediators. Journal of Physiology 498,729–734.

Ostrowski, K., Hermann, C., Bangash, A. et al. (1998a) A trauma-like elevation of plasma cytokines in humans inresponse to treadmill running. Journal of Physiology 513, 889–894.

Ostrowski, K., Rohde, T., Zacho, M., Asp,S. & Pedersen, B.K. (1998b) Evidencethat interleukin-6 is produced in humanskeletal muscle during prolongedrunning. Journal of Physiology 508,949–953.

Ostrowski, K., Schjerling, P. & Pedersen,B.K. (2000) Physical activity and plasmainterleukin-6 in humans: effect ofintensity of exercise. European Journal of Applied Physiology 83, 512–515.

Papanicolaou, D.A., Petrides, J.S., Tsigos,C. et al. (1996) Exercise stimulatesinterleukin-6 secretion: inhibition byglucocorticoids and correlation withcatecholamines. American Journal ofPhysiology, Endocrinology and Metabolism271, E601–E605.

Peake, J.M. (2002) Exercise-inducedalterations in neutrophil degranulationand respiratory burst activity: possiblemechanism of action. ExerciseImmunology Review 8, 49–100.

Pedersen, B.K. & Hoffman-Goetz, L. (2000)Exercise and the immune system:regulation, integration, and adaptation.Physiological Reviews 80, 1055–1081.

Pedersen, B.K. & Ullum, H. (1994) NK cellresponse to physical activity: possiblemechanisms of action. Medicine andScience in Sports and Exercise 26, 140–146.

Pedersen, B.K., Tvede, N., Christensen,L.D. et al. (1989) Natural killer cellactivity in peripheral blood of highly

Lymphocyte, NK and LAK cellresponses to maximal exercise.International Journal of Sports Medicine17, 60–65.

Nieman, D.C. (1997) Immune response to heavy exertion. Journal of AppliedPhysiology 82, 1385–1394.

Nieman, D.C. (1998) Exercise andresistance to infection. Canadian Journalof Physiology and Pharmacology, 76,573–580.

Nieman, D.C. (2000) Is infection risk linked to exercise workload? Medicineand Science in Sports and Exercise 32,S406–S411.

Nieman, D.C., Nehlsen-Cannarella, S.L.,Markoff, P.A. et al. (1990) The effects ofmoderate exercise training on naturalkiller cells and acute upper respiratorytract infections. International Journal ofSports Medicine 11, 467–473.

Nieman, D.C., Henson, D.A., Gusewitch,G. et al. (1993a) Physical activity andimmune function in elderly women.Medicine and Science in Sports and Exercise 25, 823–831.

Nieman, D.C., Miller, A.R., Hension, D.A.et al. (1993b) Effects of high- vs.moderate-intensity exercise on naturalkiller cell activity. Medicine and Science inSports and Exercise 25, 1126–1134.

Nieman, D.C., Ahle, J.C., Henson, D.A. et al. (1995a) Indomethacin does not alterthe natural killer cell response to 2.5 h ofrunning. Journal of Applied Physiology 79,748–755.

Nieman, D.C., Henson, D.A., Sampson,C.S. et al. (1995b) The acute immuneresponse to exhaustive resistanceexercise. International Journal of SportsMedicine 16, 322–328.

Nieman, D.C., Nehlsen-Cannarella, S.L.,Fagoaga, O.R. et al. (1998) Effects of mode and carbohydrate on thegranulocyte and monocytes response tointensive, prolonged exercise. Journal ofApplied Physiology 84, 1252–1259.

Nieman, D.C., Henson, D.A., Smith, L.L. et al. (2001) Cytokine changes after amarathon race. Journal of AppliedPhysiology 91, 109–114.

Nieman, D.C., Henson, D.A., Fagoaga,O.R. et al. (2002) Change in salivary IgAfollowing a competitive marathon race.International Journal of Sports Medicine 23,69–75.

Niess, A.M., Fehrenbach, E., Strobel, G. et al. (2003) Evaluation of stressresponses to interval training at low and moderate altitudes. Medicine andScience in Sports and Exercise 35, 263–269.

Page 382: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

366 chapter 25

trained and untrained persons.International Journal of Sports Medicine10, 129–131.

Pedersen, B.K., Steensberg, A., Fischer, C.et al. (2001) Exercise and cytokines withparticular focus on muscle-derived IL-6.Exercise Immunology Reviews 7, 18–31.

Peters, E.M. & Bateman, E.D. (1983)Ultramarathon running and upperrespiratory tract infections. Anepidemiological survey. South AfricanMedical Journal 64, 582–584.

Petrides, J.S., Gold, P.W., Mueller, G.P. et al. (1997) Marked differences infunctioning of the hypothalamic–pituitary–adrenal axis between groupsof men. Journal of Applied Physiology 82,1979–1988.

Pitzalis, C., Pipitone, N. & Perretti, M.(2002). Regulation of leukocyte-endothelial interactions byglucocorticoids. Annals of the New YorkAcadamy of Science 966, 108–118.

Pizza, F.X., Mitchell, J.B., Davis, B.H. et al. (1995) Exercise-induced muscledamage: effect on circulating leukocyteand lymphocyte subsets. Medicine andScience in Sports and Exercise 27, 363–370.

Potteiger, J.A. Chan, M.A., Haff, G.G. et al.(2001) Training status influence T-cellresponses in women following acuteresistance exercise. Journal of Strengthand Conditioning Research 15, 185–191.

Pyne, D.B. (1994) Regulation of neutrophilfunction during exercise. Sports Medicine17, 245–258.

Pyne, D.B., McDonald, W.A. Gleeson, M. et al. (2000) Mucosal immunity,respiratory illness, and competitiveperformance in elite swimmers. Medicineand Science in Sports and Exercise 33,348–353.

Rabin, B.S., Moyna, N.M., Kusnecov, A.,Zhou, D. & Shurin, M.R. (1996)Neuroendocrine effects on immunity. In: Exercise and Immune Function(Hoffman-Goetz, L., ed.). CRC Press,Boca Raton, FL: 21–37.

Rall, L.C., Roubenoff, R., Cannon, J.G. et al.(1996) Effects of progressive resistancetraining on immue response in agingand chronic inflammation. Medicine andScience in Sports and Exercise 28,1356–1365.

Ramirez, F. & Silva, A. (1997)Glucocorticoids enhance concanvalin A-induced mitogenic response throughthe inhibition of nitric oxide production.Immunology 90, 66–73.

Rhind, S.G., Gannon, G.A., Suzui, M.,Shephard, R.J. & Shek, P.N. (1999)Indomethacin inhibits circulating PGE2

to excessive stress? Medicine and Sciencein Sports and Exercise 32, 317–331.

Smith, L.L. & Miles, M.P. (2000) Exercise-induced muscle injury andinflammation. In: Exercise and SportScience (Garrett, W.E. & Kirkendall, D.T.,eds.). Lippincott, Williams & Wilkins,Philadelphia, PA: 401–411.

Smith, L.L., Anwar, A., Fragen, M. et al.(2000) Cytokines and cell adhesionmolecules associated with high-intensityeccentric exercise. European Journal ofApplied Physiology 82, 61–67.

Smith, J.A., Telford, R.D., Baker, M.S.,Hapel, A.J. & Weidemann, M.J. (1992)Cytokine immunoreactivity in plasmadoes not change after moderateendurance exercise. Journal of AppliedPhysiology 73, 1396–1401.

Smits, H.H., Grunberg, K., DeRijk, R.H.,Sterk, P.J. & Hiemstra, P.S. (1998)Cytokine release and its moduclation by dexamethasone in whole bloodfollowing exercise. Clinical andExperimental Immunology 111, 463–468.

Spangenburg, E.E. & Booth, F.W. (2002)Multiple signaling pathways mediateLIF-induced skeletal muscle satellite cell proliferation. American Journal ofPhysiology 283, C204–C211.

Sprenger, H., Jacobs, C., Nain, M. et al.(1992) Enhanced release of cytokines,interleukin-2 receptors, and neopterinafter long-distance running. ClinicalImmunology and Immunopathology 63,188–195.

Steensberg, A., Toft, A.D., Bruunsgaard, H.et al. (2001) Strenuous exercise decreasesthe percentage of type 1 T cells in thecirculation. Journal of Applied Physiology91, 1708–1712.

Steensberg, A., Keller, C., Starkie, R.L. et al.(2002) IL-6 and TNF-α expression in, andrelease from, contracting human skeletalmuscle. American Journal of Physiology283, E1272–E1278.

Steensberg, A., Fischer, C.P., Keller, C.,Møller, K. & Pedersen, B.K. (2003) IL-6enhances plasma IL-1ra, IL-10, andcortisol in humans. American Journal ofPhysiology, Endocrinology and Metabolism285, E433–E437.

Steinacker, J.M., Lormes, W., Reissnecker,S. & Liu, Y. (2004) New aspects of thehormone and cytokine response totraining. European Journal of AppliedPhysiology 91, 382–391.

Strasner, A., Davis, J.M., Kohut, M.L. et al.(1996) Effects of exercise intensity onnatural killer cell activity in women.International Journal of Sports Medicine 18,56–62.

and reverses postexercise suppression of natural killer cell activity. AmericanJournal of Physiology 276, R1496–R1505.

Riccardi, C., Bruscoli, S. & Micliorati, G.(2002) Molecular mechanisms ofimmunomodulatory activity ofglucocorticoids. Pharmacological Research45, 361–368.

Rivest, S. (2001) How circulating cytokinestrigger the neural circuits that controlthe hypothalamic–pituitary–adrenalaxis. Psychoneuroendocrinology 26,761–788.

Rogers, M.A., Stull, G.A. & Apple, F.S.(1985) Creatine kinase isoenzymeactivities in men and women following a marathon race. Medicine and Science inSports and Exercise 17, 679–682.

Ronsen, O., Kjeldsen-Kragh, J., Haug, E.,Bahr, R. & Pedersen, B.K. (2002a)Recovery time affects immunoendocrineresponses to a second bout of enduranceexercise. American Journal of Physiology,Cellular Physiology 283, C1612–C1620.

Ronsen, O., Lea, T., Bahr, R. & Pedersen,B.K. (2002b) Enhanced plasma IL-6 andIL1ra responses to repeated vs. singlebouts of prolonged cycling in eliteathletes. Journal of Applied Physiology 92,2547–2553.

Sanders, V.M., Baker, R.A., Ramer-Quinn,D.S. et al. (1997) Differential expressionof the β2-adrenergic receptor by Th1 andTh2 clones: implications for cytokineproduction and B cell help. Journal ofImmunology 158, 4200–4210.

Schedlowski, M., Falk, A., Rohne, A. et al. (1993) Catecholamines inducealterations of distribution and activity of human natural killer (NK) cells.Journal of Clinical Immunology 13,344–351.

Schedlowski, M., Hosch, W., Oberbeck, R.et al. (1996) Catecholamines modulatedhuman NK cell circulation and functionvia spleen-independent β2-adrenergicmechanisms. Journal of Immunology 156,93–99.

Selye, H. (1936) A syndrome produced bydiverse nocuous agents. Nature 138,32–36.

Semark, A., Noakes, T.D., St. Clair Gibson,A. & Lambert, M.I. (1999) The effect of a prophylactic dose of flubiprofen on muscle soreness and sprintingperformance in trained subjects. Journalof Sport Science 17, 197–203.

Shephard, R.J. (1997) Physical Activity,Training, and the Immune Response.Cooper Publishing Group, Carmel, IN.

Smith, L.L. (2000) Cytokine hypothesis ofovertraining: a physiological adaptation

Page 383: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

immune response to exercise and doms 367

Stupka, N., Tarnopolsky, M.A., Yardley,N.J. & Phillips, S.M. (2001) Cellularadaptation to repeated eccentricexercise-induced muscle damage.Journal of Applied Physiology 91,1669–1678.

Suzuki, K., Sato, H., Kikuchi, T. et al. (1996)Capacity of circulating neutrophils toproduce reactive oxygen species afterexhaustive exercise. Journal of AppliedPhysiology 81, 1213–1222.

Suzuki, K., Totsuka, M., Nakaji, S. et al.(1999). Endurance exercise causesinteraction among stress hormones,cytokines, neutrophil dynamics, andmuscle damage. Journal of AppliedPhysiology 87, 1360–1367.

Suzuki, K., Nakaji, S., Yamada, M. et al.(2002) Systemic inflammatory responseto exhaustive exercise. Cytokinekinetics. Exercise Immunology Review 8,6–48.

Tharp, G.D. & Barnes, M.W. (1990)Reduction of salivary immunoglobulinlevels by swim training. European Journalof Applied Physiology 60, 61–64.

van Tits, L.J.H., Michel, M.C., Grosse-Wilde, H. et al. (1990) Catecholaminesincrease lymphocyte β2-adrenergicreceptors via a β2-adrenergic, spleen-dependent process. American Journal ofPhysiology 258, E191–E202.

Toft, A.D., Jensen, L.B., Bruunsgaard, H. et al. (2002) Cytokine response to

ubiquitin expression after repeatedeccentric exercise. Medicine and Science in Sports and Exercise 35, 2023–2031.

Woods, J.A. & Davis, J.M. (1994) Exercise,macrophages, and cancer defense.Medicine and Science in Sports and Exercise26, 147–157.

Woods, J.A. Davis, J.M., Mayer, E.P.,Ghaffar, A. & Pate, R.R. (1994) Effects ofexercise on macrophage activation forantitumor cytotoxicity. Journal of AppliedPhysiology 76, 2177–2185.

Woods, J.A., Ceddia, M.A., Kozak, C. &Wolters, B.W. (1997) Effects of exerciseon the macrophage MHC II response toinflammation. International Journal ofSports Medicine 18, 483–488.

Woods, J.A., Davis, J.M., Smith, J.A. &Nieman, D.C. (1999) Exercise andcellular innate immune function.Medicine and Science in Sports and Exercise31, 57–66.

Yamada, M., Suzuki, K., Kudo, S. et al.(2002) Raised plasma G-CSF and IL-6after exercise may play a role inneutrophil mobilization into thecirculation. Journal of Applied Physiology92, 1789–1794.

Zhou, J., Olsen, S., Moldovan, J. et al. (1997)Glucocorticoid regulation of naturalcytotoxicity: effects of cortisol on thephenotype and function of clonedhuman nature killer cell line. CellularImmunology 178, 108–116.

eccentric exercise in young and elderlyhumans. American Journal of Physiology283, C289–C295.

Tsigos, C. & Chrousos, G.P. (2002)Hypothalamic–pituitary–adrenal axis,neuroendocrine factors and stress.Journal of Psychosomatic Research 53,865–871.

Turnbull, A.V., Dow, R.C., Hopkins, S.J. et al. (1994) Mechanisms of activation of the pituitary-adrenal axis by tissue injury in the rat.Psychoneuroendocrinology 19, 165–178.

Urhausen, A. Gabriel, H. & Kindermann,W. (1995) Blood hormones as markers oftraining stress and overtraining. SportsMedicine 20, 251–276.

Walsh, N.P., Bishop, N.C., Blackwell, J.,Wierzbicki, S.G. & Montague, J.C. (2002)Salivary IgA response to prolongedexercise in a cold environment in trainedcyclists. Medicine and Science in Sports andExercise 34, 1632–1637.

Warhol, M.J., Siegel, A.J., Evans, W.J. &Silverman, L.M. (1985) Skeletal muscleinjury and repair in marathon runnersafter competition. American Journal ofPathology 118, 331–339.

Weicker, H. & Werle, E. (1991) Interactionbetween hormones and the immunesystem. International Journal of SportsMedicine 12 (suppl. 1), S30–S37.

Willoughby, D.S., Taylor, M. & Taylor, L.(2003) Glucocorticoid receptor and

Page 384: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

368

Introduction to exercise–neuroendocrine–immune relationships

The many benefits of regular physical exercise arenot only well-documented in the scientific literaturebut have also received considerable attention in thepopular press. It is recognized that exercise haswide-ranging effects on most organ systems of thebody but, in general, emphasis has been placed onits impact on cardiovascular and pulmonary func-tion (reviewed in Booher & Smith 2003). However,more recently, the cellular and molecular mechan-isms underlying the effects of exercise on severalother organs systems, especially the immune sys-tem, have begun to be delineated (e.g. Pedersen & Hoffman-Goetz 2000; Pedersen & Toft 2000;Suzuki et al. 2002; Woods et al. 2002; Lakier-Smith2003; Nieman 2003). The strong interest in the rela-tionship between exercise and the immune functionhas lead to the recent publication of a number ofexcellent review articles devoted to this subject(International Journal of Sports Medicine [volume 21,supplement 1, May 2000]; Immunology and Cell Bio-logy [volume 78, October 2002]).

It has long been recognized that exercise can havea strong impact on a person’s overall mental condi-tion as well as their physical state (Glenister 1996;Fox 1999; Paluska & Schwenk 2000; Salmon 2001).Of particular recent interest is how one’s mentalstate can, in turn, influence a wide range of phy-siological parameters that together contribute to maintaining homeostasis and overall well-being.Although the physiological benefits of exercise havebeen recognized for quite some time, the contribu-

tions of exercise to physiology from a psychologicalperspective are less well established. The exactmechanisms underlying these contributions havenot been identified; but it is generally accepted thatthe functioning of both the nervous and endocrinesystems are intimately associated with a variety ofpsychological states. The fact that exercise can affectcomponents of both the nervous and endocrine systems suggests that, in part, these effects may bemediated via a psychological pathway. In recentyears, substantial evidence has been collected tosupport a functional link between not only the nervous and endocrine systems but also among thenervous, endocrine and immune systems (Fig. 26.1)(Conti et al. 2000; Ader et al. 2001).

Given the many findings that both the nervousand endocrine systems are alone able to modu-late a variety of immune functions provides strong

Chapter 26

The Impact of Exercise on Immunity: the Role ofNeuroendocrine–Immune Communications

ANDREA M. MASTRO AND ROBERT H. BONNEAU

Nervoussystem

Endocrinesystem

Immunesystem

Exercise

Fig. 26.1 Exercise and the nervous–endocrine–immunenetwork.

Page 385: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 369

support for a psychological link between exerciseand immunity. As is outlined below, there is indeeda neuroendocrine-mediated link between exerciseand immune function. This link may play an import-ant role in the development and progression of diseases that are either immunologically resisted(e.g. infectious diseases, cancer) or that are causedby an undesirable activation of the immune system(e.g. allergy, autoimmunity).

It is important to note that the information providedin this chapter is not intended to represent a compre-hensive review of all literature dealing with exerciseand immunity. For such information, the reader isdirected to several recent reviews in this area(Hoffman-Goetz 1996; Nieman & Pedersen 2000;Pedersen & Hoffman-Goetz 2000; Shephard & Shek2000a; Hoffman-Goetz & Pedersen 2001). Rather,this chapter is specifically designed to enlighten thereader on the neuroendocrine-mediated mechan-isms underlying the relationship among exerciseand specific aspects of immune function. It is alsointended to provide insight into the areas in whichan understanding of the relationship between exer-cise and immune function can be further expandedas a result of recent advances in our knowledge ofimmune function and state-of-the-art experimentalapproaches that can quantify such function. Lastly,we will address the potential impact of exercise ondiseases whose prevention or cause is associatedwith one or more aspects of immune function.

Neuroendocrine–immunecommunications

Neuroendocrine communications

Before launching into a discussion of the variety of functional relationships that exist between theneuroendocrine and immune systems and theirimpact on one’s level of immunocompetence, it isfirst necessary to briefly review some basic prin-ciples underlying the communications that existbetween the nervous and endocrine systems andhow these communications are possibly influencedby exercise. However, it should be noted that a com-prehensive review of the both the nervous andendocrine systems, as well as exercise physiology, is

clearly beyond the scope of this text and is discussedin depth elsewhere (Robergs & Keteyian 2003).

the lines of communication between

the nervous and endocrine systems

It has been clear for quite some time that nervousand endocrine systems function as a single inter-related system within the body. The functional rela-tionships that exist between these two systems andhow they regulate a wide range of bodily processesis the basis of the study of neuroendocrinology. Thisrelationship operates bi-directionally in that theendocrine system affects the nervous system andthe nervous system affects the endocrine system.Whereas hormones serve as the primary mediatorsof the endocrine-to-nervous system communica-tion, the nervous system–endocrine communicationoccurs within the site of direct interface, at the levelof the neuroendocrine cell itself.

Hormones impact virtually every tissue in thebody as a result of their transport through the blood.Functional interactions between hormones andtheir target tissues are highly dependent on specificreceptor-mediated binding of hormones to the cellswhich are able to respond. Depending on the hor-mone, these receptors are located on either the cellmembrane, within the cytoplasm, or in the nucleus.The mechanisms of hormone action are varied andinclude membrane transport, stimulation of genetranscription, and the activation of intracellular second messengers such as cyclic adenosine mono-phosphate (cAMP).

There are a variety of mechanisms that regulatethe release of hormones into the neuroendocrinepathway. Endogenous circadian or diurnal rhythmsof release provide a tonic cyclic pattern of hormonesecretion that is independent of physiologic pertur-bation. Superimposed on these endogenous rhythmsare a number of highly complex positive and negat-ive feedback loop mechanisms which help to main-tain endocrine-associated homeostasis.

function and importance of

neuroendocrine communications

Neuroendocrine connections are important in that

Page 386: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

370 chapter 26

they allow our bodies to maintain a state of home-ostasis. For example, neuroendocrine relationshipsplay a critical role in water conservation and main-tenance of body fluid osmolarity, blood volume andpressure, growth and development, metabolism,electrolyte balance, ovulation and parturition andbehavior. Disorders in the generation, secretion, orresponse to hormones can lead to a wide range ofpathological conditions including diabetes insipidus,osteoporosis and acromegaly, to name a few.

The neuroendocrine system also provides adapt-ive physiological responses which allow one torespond to and cope with changes in the environ-ment. This ability to adapt to such changes is centralto maintaining a number of physiological parameterswithin a ‘normal range’ that allows for survival. For example, the hypothalamic–pituitary–adrenal(HPA) axis is among the first physiological responsesystems to become activated in response to environ-mental ‘stress’. Although ‘stress’ can be thought offrom either a strictly psychological or physiologicalstandpoint, or a combination of both, the ultimatebodily response is the activation of the HPA axis(Chrousos & Gold 1992; Dhabhar & McEwen 2001).

A well-orchestrated communication between thecells and tissues comprising the HPA axis is essen-tial in maintaining homeostasis under conditions ofstress. Signals from the limbic system trigger therelease of corticotrophin-releasing hormone (CRH)from the paraventricular nucleus of the hypotha-lamus which, in turn, induces the anterior pituitaryto release adrenocorticotropic hormone (ACTH).ACTH then enters the circulation where it interactswith cells of the adrenal cortex to produce cortisol in humans (corticosterone in rats and mice). It isimportant to recognize that through the ability ofeach of these compounds to feedback onto theorgans in which they were made, the synthesis ofthese molecules is well-controlled.

neuroendocrine adaptations

to exercise

The ability of the body to tolerate exercise is regu-lated by complex interactions between the auto-nomic nervous system and the endocrine system.The body responds to both neural stimulation and

specific chemical and mechanical conditions thathelp to regulate, through the action of a variety ofhormones, a number of physiological functions during exercise. Some of these functions includeenergy metabolism, fuel mobilization, fluid balance,vascular hemodynamics and protein synthesis.These responses can vary among individuals and beinfluenced by both exercise intensity and gender.

The exercise-induced, hormonally-based regula-tion of physiological processes involves a number of hormones including cortisol, growth hormone,vasopressin (antidiurectic hormone), renin, aldos-terone, thyroxin, insulin, glucagons, and the cate-cholamines, epinephrine and norepinephrine. Theepinephrine and norepinephrine released from theadrenal gland control changes in muscle metabol-ism, cardiac output and vascular resistance. Theremay also be changes in the levels of other hormones(e.g. estrogen, follicle-stimulating hormone [FSH],luteinizing hormone [LH] and testosterone, α- andβ-endorphins and enkephalins) that are not neces-sarily associated with the maintenance of homeo-stasis. Peptide hormones such as growth hormone,insulin and prolactin increase with exercise as partof the metabolic response. Many, if not all, of thesehormones are able to bind to immune cells and elicita variety of cellular responses.

The above mentioned neuroendocrine responsesto exercise have been recognized for quite sometime. In addition, many of the hormones that are the products of these responses have been shown to affect various aspects of immune function both in vitro and in vivo in the context of studies totallyunrelated to exercise. However, as outlined in depthlater this chapter, it is now recognized the exerciseitself may indeed influence immunity through theactions of these neuroendocrine-derived hormones.

Nervous–endocrine–immune communications

introduction to neuroimmunology

As is briefly outlined above, intricate levels of com-munication exist between nervous and endocrinesystems. In fact, most the other major organs systemsof the body (e.g. circulatory, respiratory, gastroin-testinal, reproductive) are functionally linked with

Page 387: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 371

both the nervous and endocrine systems and/orwith each other in a number of ways. However,from a historical perspective, the immune systemhad generally been thought to function autonom-ously with little or no input from any of the otherorgan systems. However, during the past threedecades there have a plethora of both human(Solomon & Moos 1964; Solomon et al. 1966; Solomon1981a, 1981b; reviewed in Glaser & Kiecolt-Glaser1994) and animal (Solomon et al. 1968; Solomon1969; Ader & Cohen 1975; reviewed in Bonneau et al.2001; Moynihan & Stevens 2001) studies that haveprovided substantial evidence that the immune sys-tem is functionally integrated with both the centralnervous system (CNS) and with the endocrine sys-tem. Given the fact that the identification of many ofthe complex interactions among these three systemshad their roots in psychology, this area of study hastraditionally been known as ‘psychoneuroimmuno-logy’ (Greer 2000) although a simpler term, ‘neuro-immunology’ may be equally appropriate.

Defining the relationship among the nervous,endocrine, and immune systems is not easy for several reasons. First and foremost, the immune system itself is very complex. It is comprised of bothprimary (bone marrow and thymus) and secondary(lymph nodes, spleen) immune organs distributedthroughout the body. Although for practical andethical considerations blood-derived immune cellsare studied most often in humans, one can not forgetthe fact that there are immune cells in other parts ofthe body that play important regulatory roles. Forexample, a variety of immune-derived cells lineboth the respiratory and intestinal systems and con-tribute to what is known as mucosal immunity.Together with the intraepithelial lymphocytes whichreside in the skin, these cells provide an importantfirst line of defense against invading pathogenicmicroorganisms. Thus, by limiting studies to onlyblood-borne immune cells we may compromise our ability to truly learn the importance of neuro-endocrine–immune interactions in overall immune-mediated defense. A second difficulty in definingthe neuroendocrine–immune relationship is thatour understanding of immune function continues togrow at a rapid pace, independent of any additionalknowledge of the nervous and endocrine systems.

At the cellular level, much more is known about thefunctioning of both the innate and adaptive arms of the immune system and how these two arms are intertwined via the synthesis of and response to cytokines. A better understanding of immuneevents at the molecular levels have been advancedby developments in molecular biology and throughthe use of transgenic animals. However, our under-standing of how the neuroendocrine systems affectthese molecular events is just beginning to bedefined.

mediators and mechanisms of

neuroendocrine–immune interactions

Recent experimental evidence has proven that theCNS–endocrine–immune axis (neuroendocrine–immune axis) operates in a bi-directional fashion(Chambers et al. 1993; Felten et al. 1993; Moynihan & Ader 1996; Stevens-Felten & Bellinger 1997). Thatis, the immune system receives signals from the nervous system and the immune system providesinformation to the nervous system. This intercel-lular communication can be mediated by productsof the immune system including cytokines, growthfactors and even neuropeptides made by the lym-phocytes themselves. Therefore, the distinctions thathad once been made among lymphokines, growth,factors, hormones and neuropeptides with respectto the organ system in which they function are nolonger appropriate.

Studies of the relationship between neuro-peptides and the regulation of immune functionhave focused particularly on those neuropeptidesderived from the polyprotein proopiomelanocortin(POMC), particularly ACTH and β-endorphin.Other hormones such as cortisol, growth hormone,prolactin and the catecholamines, epinephrine and norepinephrine are also central to our under-standing of the relationship between endocrine and immune function. Overall, the evidence forneuronal, endocrine and immune intersystem com-munications have formed the basis of numerousstudies that have investigated the relationshipamong physical stressors such as exercise, immunefunction and the health status of an individual (Fig. 26.2).

Page 388: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

372 chapter 26

The ability of the immune system to respond toneuroendocrine-derived peptides and hormonesdepends on the presence of functional receptors onthe immune cells themselves. Indeed, there are avariety of immune-derived cells (e.g. lymphocytesand monocytes) that possess receptors for neuro-endocrine-derived peptides and hormones that areidentical to those receptors that are present on cellsof the nervous and endocrine systems (Weigent et al.1990; Blalock 1994; DeKloet et al. 1994; Garza & Carr1997). For example, catecholamines, opioids, sero-tonin, vasopressin, ACTH, growth hormone andprolactin can all influence various aspects of theimmune system though receptor-specific binding.Likewise, there are receptors on cells of the neuro-endocrine system for immune-derived productssuch as cytokines.

In order for specific receptors to provide a linkbetween the neuroendocrine and immune systems,the neuroendocrine-derived products must firstcome in contact with the cells of the immune system.Some products are released into the blood and cir-culate to the immune cells that are located through-out the body. Alternatively, products of the nervoussystem may be released from nerve terminals in the direct proximity of immune cells located in theprimary (thymus) and secondary (lymph nodes,spleen) lymphoid tissues. The latter mechanism issupported by the finding that nerve fibers of theautonomic nervous system directly innervate theprimary and secondary lymphoid tissues (Livnat

et al. 1985; Felten & Felten 1988; Bellinger et al. 1992;Madden et al. 1995) in much the same way that otherorgans of the body, such as the heart, are innervatedby the fibers of nervous system. This innervation ofthe thymus, lymph nodes and spleen is important inoverall immune function given that these are sites of lymphocyte development and antigen-specificlymphocyte activation. Such innervation had beenreferred to as a type of ‘hard wiring’ of the nervoussystem with the immune system.

function of neuroendocrine–immune

connections

The bi-directional levels of communication amongthe cells, tissues and organs that comprise the nerv-ous, endocrine and immune systems suggests thatthe overall control and function of these systems ismuch more complex than had been once thought.Thus, the physiological processes that are critical inmaintaining homeostasis and well-being under anumber of environmental insults can theoreticallybe controlled in many ways.

A series of well-orchestrated events at both thecellular and molecular level are necessary for theproper functioning of the immune system. His-torically, the orchestration of these events has beengenerally recognized to be mediated by the varietyof cell types that comprise the immune system itself. Cells such as B-lymphocytes, cytotoxic T-lymphocytes, helper T-lymphocytes, macrophages,neutrophils, dendritic cells and natural killer (NK)cells are the most common. In recent years, theidentification of the many molecules that each ofthese cell types synthesize (e.g. cytokines, chemo-kines, growth factors, etc.) and their critical role in the regulation of overall immune function hasallowed for a better understanding of the immuneresponse at both the cellular and molecular levels.However, cells and molecules of the nervous andendocrine systems can also be key contributors togeneral immune function, thus making it moredifficult to fully understand what controls the func-tioning of the immune system.

To date, it has been shown that nearly every celltype and function within the immune system can be modulated by products of the nervous and

Homeostasis

CytokinesNeuroendocrine

hormonesNeuroendocrine

systemsImmunesystem

Disease

Fig. 26.2 Health and disease. (Adapted from Chambers &Schauenstein 2000.)

Page 389: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 373

endocrine systems. A comprehensive review of thisliterature is beyond the scope of this chapter.However, some broad examples include neuroen-docrine effects on antigen processing and presenta-tion, antibody production, lymphocyte activationand proliferation, cytokine production and NK celllytic activity. These effects are mediated at the levelof the ligand–receptor interaction, second messengersystems and gene expression. Events such as exer-cise and stress contribute to these effects by theirability to induce the synthesis of products of the nervous and endocrine systems.

The immune system is intimately involved in ourwell-being and survival. The ability to ward off thedeleterious consequences of infectious pathogensmay depend on the generation of an immune res-ponse following vaccination either during child-hood (poliovirus, hepatitis virus) or as an adult(influenza virus). For infectious pathogens forwhich no vaccinations currently exist, our ability to mobilize an effective innate immune responsethrough the activation of immune cells such as neutrophils, NK cells and macrophages is essential.For some pathogens, the further development of anadaptive immune response, a response targetedspecifically toward the challenge pathogen, may becritical. Such a response is typically mediated by theB-lymphocytes which produce antibodies and theT-lymphocytes which help destroy those cells thatthe pathogen has invaded and relies on for its pro-pagation and continued insult on the body. Lastly,our ability to generate and maintain immunologicalmemory, as a consequence of vaccination and/oractual infection, is also important in our long-termdefense against infection. The fact that products ofthe nervous and endocrine systems affect all of theabove underscores the important role of both thenervous and endocrine systems in the defenseagainst infectious pathogens.

There is also substantial interest in the role of theimmune system in the development and progres-sion of cancer. Although the precise contributions of the various components of the immune system inthe defense against cancer have not yet been fullydefined, there is substantial evidence to believe thatimmune components such as NK cells and cytotoxicT-lymphocytes (CTL) may indeed play a key role.

Recently, there has been experimental data to sup-port the notion that products of the neuroendocrinesystem can contribute to the immune system’s rolein the defense against cancer (Berczi et al. 1998; Ben-Eliyahu & Shakhar 2001; Turner-Cobbs et al. 2001;Sephton & Spiegel 2003). The successful defenseagainst infection and cancer relies on an enhancedlevel of immune function. In contrast, it may bedesirable to suppress immune function in order toreduce the severity of diseases that result as a con-sequence of an overactive or inappropriately tar-geted immune response. A number of autoimmunedisorders fall into this category and include well-known diseases such as rheumatoid arthritis, juven-ile diabetes, systemic lupus erythematosus (SLE)and scleroderma. Likewise, it is desirable to dimin-ish the magnitude of the immune responses that are involved in mediating allergic reactions and thatare responsible for the graft rejection in organ transplant recipients. Exactly how products of theneuroendocrine system contribute to each of theseresponses is a subject of much interest.

It is important to note that our knowledge of the inner workings of the immune system hasrapidly involved during the past three decades.Advances in many areas of biological technologyhave allowed for a substantially better understand-ing of immune function at both the cellular and themolecular levels. Concomitantly, there has been anincrease in our knowledge of the role of the nerv-ous and endocrine systems at each of these levels, thus advancing the field of neuroimmunology andfurthering our understanding of neuroendocrine–immune interactions.

The impact of exercise-induced effectson the immune system

Exercise and immunity

‘Exercise helps relieve fatigue and other symptomsof disease’, proclaimed a recent newspaper head-line. The article (Bertrand 2003) described how exer-cise helped individuals cope with diseases such as multiple sclerosis and cancer. It included a‘checklist’ of approaches to developing an exerciseregimen. For example, there were suggestions of

Page 390: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

374 chapter 26

how to monitor pulse and breathing as well as howto pace activity or to modify the intensity and dura-tion of exercise to meet one’s needs. Similar articlesrelating exercise to good health and immunity arecommon in popular journals, self-help books andnewspapers. While it is generally accepted thatsome physical activity is better than no activity andthat exercise is ‘good for you’, exactly how exer-cise impacts on the immune system is not clear.Although one researcher in the field of exercise andimmunity rather skeptically stated that, ‘. . . num-erous attempts to link exercise to meaningful alterations in immune function have been largelyunconvincing (Moseley 2000, p. 128), the potentialimpact of exercise on immune function can not beignored nor should studies of such an impact be terminated. This is especially true in light of ourincreasing knowledge of immunity and the moresophisticated means of quantifying immune function.

The quest to understand the connections betweenexercise and health is driven by several perspect-ives (Mackinnon 2000a). If exercise can be used totreat individuals with disease, can exercise prevent disease? On the other hand, can a person exercisetoo much and actually increase susceptibility to disease? For example, some athletes undergoingintensive and long duration exercise training sufferfrom upper respiratory tract infections (URTIs).Does this observation imply that exercise caninduce immunosuppression and thereby increasethe risk of infection? Can exercise protect one fromor exacerbate autoimmunity? What happens to theimmune system of either bedridden patients whoare largely inactive or astronauts while in space?Does moderate exercise modulate the immune sys-tem? Questions regarding the relationship betweenphysical activity and good health have been askedfor centuries, but the mechanisms by which exer-cise relates to immunity have only seriously beenstudied in a systematic approach for the past 30years or so.

Coupled with the complexity of the immune sys-tem is the complexity of the exercise protocol that is being studied. The potential impact of exercisehas much to do with the type, mode, intensity andduration of the exercise. The effect of exercise onimmunity may also be further confounded by age,

gender and prior fitness level among other variablesin the humans being studied. Many studies havebeen carried out using animal models of exerciseand immunity. Although such studies have pro-vided valuable information regarding this relation-ship, the translation of this information to thehuman condition is not always straightforward. For the most part, the information discussed in thischapter has been limited to that which has beenobtained in human studies.

Evaluating the impact of exercise on the immunesystem in humans

Important in interpreting how exercise affects theimmune system are the methodologies that are usedto assess immune function. Two commonly usedassays in human subject studies are the determina-tion of the subpopulation of the blood leukocytes(phenotyping) and ex vivo stimulation of lympho-cytes in culture (lymphocyte activation). Due toboth convenience and ethical considerations, mosthuman studies rely exclusively on peripheral bloodas source of lymphocytes for analysis. However,blood contains only 1–2% of the immune cells in the body, and many of these cells are constantlytrafficking throughout the body, entering and exit-ing various sites where infectious pathogens arecommonly encountered. Typically, flow cytometryis used to enumerate the populations of these cells inthe blood. This flow cytrometric approach makesuse of fluorescently tagged monoclonal antibodiesthat bind to specific cell surface proteins known as CD, or cluster of differentiation, markers. Thesesurface proteins are used to differentiate and quant-itate the various cell types of the immune system.For example, CD3+/CD4+ cells are T-helper cellswhile CD3+/CD8+ cells are T-cytotoxic cells. How-ever, a change in the distribution and number ofthese cells in blood does not necessarily indicate a change in cell function nor does such a changereflect an immune response at some other locationin the body.

A variety of approaches have been used to studyimmune cell function. One commonly used tech-nique is ex vivo culture of lymphocytes in the pres-ence of compounds such as concanavalin A or

Page 391: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 375

phytohemagglutinin, polyclonal T-cell mitogenswhich stimulate the lymphocytes to produce a vari-ety of cytokines, to express receptors and to divide.Although this technique is fairly straightforward,the results are subject to a variety of experimentalmanipulations and can be affected by the particu-lar combination of the immune-derived cells that are located in the blood sample being tested. Toallow for changes in cell numbers after exercise, theresulting functional data are often mathematicallynormalized to the number of T-cells in the sample;however, the presence of other cell types in theblood following exercise also can affect the magni-tude of T-lymphocyte activation.

The information that can be gained from using the above techniques are necessarily limited by thesource of the lymphocyte sample and the fact thatmeasures of lymphocyte number and activationpotential in vitro are fairly general and may notaccurately reflect what is occurring in vivo. Newertechnologies such as the ability to quantify markersof cellular activation and cytokine synthesis by cellsdirectly ex vivo offer the promise of unraveling themechanisms underlying exercise-induced effects onimmune responsiveness.

Acute exercise as a stressor

As both noted above and described in detail below,exercise is known to have many beneficial effects onthe body, including the immune system. However,depending on whether or not a given exercise is perceived as a stressor by the body will determineits impact on immune function. The impact of both physical and psychological stressors on theimmune system has been studied in great depth andhas been reviewed in detail elsewhere (Glaser &Kiecolt-Glaser 1994; Buckingham et al. 1997; Rabin1999; Marsland et al. 2002; Moynihan 2003; Padgett& Glaser 2003). Until recently, all forms of stresswere thought to be generally immunosuppressive.However, it is now recognized that both the typeand degree of stress can determine its ultimateimpact on the immune system. For example, if astressor is perceived by the body as a ‘negative stressor’ (distress), then both the type and degree ofneuroendocrine activation may result in immuno-

suppression. However, if the stressor is perceived as a ‘positive stressor’ (eustress), then the neuro-endocrine-mediated effects may actually result inimmunoenhancement (Dhabhar & McEwen 2001).Thus, in establishing associations between exerciseand the immune function, one has to consider notonly whether or not the exercise is even perceived asa stressor but also if the stressor is one which resultsin immunosuppression or immunoenhancement.

In general, in studies of the relationship betweenexercise and immunity, researchers consider a boutof acute exercise as a model of induced stress ortrauma while a long duration, high intensity exer-cise is used as a way to bring about immunosup-pression (Hoffman-Goetz & Pedersen 1994, 2001).The response of the immune system to a bout ofacute exercise has been compared to that of traumaor surgery (an example of an acute bout of exerciseis 60 min on a treadmill or bicycle). Following thisexercise, numerous changes have been observedincluding increased leukocyte mobilization, releaseof proinflammatory and anti-inflammatory cyto-kines, tissue damage, production of free radicalsand the activation of some of the pathways seen ininflammation such as acute phase response, coagu-lation and fibrinolysis. These observations had ledsome to conclude that muscle damage associatedwith exercise initates an inflammatory responsewhich is responsible for these immune changes(Hoffman-Goetz 1996). However, it is now knownthat muscle damage is not essential for many ofthese changes in immune parameters (Pedersen &Hoffman-Goetz 2000). Nevertheless, when muscledamage does occur, the immune system, especiallythe innate system, can effectively bring about heal-ing. There are many changes that occur following about of acute exercise regardless of muscle damage.For example, a general leukocytosis (i.e. a several-fold increase in circulating white blood cells) is acommon finding. In exercise-induced leukocytosis,all of the white blood cell populations usuallyincrease to some extent. However, cells associatedwith the innate immune system such as neutrophils,NK cells and monocytes increase more dramatic-ally than do lymphocytes, the subclass of leukocytesthat is responsible for the acquired immune res-ponse. Within the lymphocyte population, the CD8+

Page 392: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

376 chapter 26

T-lymphocytes (cytotoxic T-lymphocytes) increasemore than do the CD4+ T-lymphocytes (T-helper);B-lymphocyte numbers typically change less. Leu-kocytosis is a rapid and transient event. It occurswithin about 30 min following the end of the exer-cise session but, after a few hours into the recoveryphase, cell numbers usually return to baseline.Sometimes there is a biphasic response in whichthere is a decline in leukocyte numbers followed by a second increase. Because of the rapidity of theincrease in leukocytes, it is believed that hemody-namics play the major role in recruiting cells fromintravascular marginated pools and storage sitessuch as the spleen. Cortisol and catecholamines,hormones that are both intimately associated withstress, are believed to be involved in this cell recruit-ment (Mackinnon 2000a). Moreover, the recruitedlymphocytes tend to be the memory cells (CD45RO+)in contrast to newly-differentiated cells (CD45RA+)(Gabriel et al. 1993). Memory cells are those whichhave previously responded to foreign material(antigen) and are programmed to quickly respondto another encounter with the same antigen.Whereas the newly differentiated cells tend to bepredominantly located in lymphoid organs, thememory cells tend to home to tissues as well as tolymphoid organs. Thus, this somewhat selectiveleukocytosis of the two populations which reside in different lymphoid compartments may provideclues to the hemodynamic and hormone drivenincreases in these cells in the blood. How these exer-cise-induced, hormone-mediated changes in leuko-cytosis influences immune responses to pathogensis of significant interest.

An intense bout of acute exercise also affects the endocrine system by elevating the levels of variety of hormones such as the catecholamines(epinephrine, norepinephrine), growth hormone, β-endorphins, sex steroids and cortisol (Hoffman-Goetz & Pedersen 2001) in the blood. Thus, giventheir location it seems logical that these hormonescould also have an influence on immune celltrafficking. In support of this hypothesis, the levelsof expression of β-adrenergic receptors on the whiteblood cells roughly correlates with the increase inthe cell numbers seen following exercise (Hoffman-Goetz & Pedersen 2001). Leukocytes have recep-

tors for all of these molecules; i.e. catecholamines,growth hormone, β-endorphins, sex steroids andcortisol, and each of these hormones administeredindividually has been reported to affect lymphocytetrafficking. These data generally support a model in which catecholamines bind to the β-adrenergicreceptors to bring about the immediate acute effectswhile corticosteroids play a more important role in exercise of greater duration (Hoffman-Goetz &Pedersen 2001).

Exercise may also bring about an increase in circulating levels of compounds that are agents ofapoptosis (programmed cell death) for lympho-cytes. For example, increased levels of cortisol andreactive oxygen species (ROS) could cause lympho-cyte apoptosis which would contribute to loss oflymphocytes from the circulation. However, in astudy designed to test this hypothesis, it was notedthat increases in cortisol, F2-isoprostanes (indic-ative of ROS), epinephrine and norepinephrineincreased with exercise but the total number of cir-culating apoptotic lymphocytes did not (Steensburget al. 2002). This finding does not rule out the importance of these molecules but suggests thatthey may not necessarily influence circulating bloodcell numbers.

lymphocyte subpopulations

T-lymphocytes are an essential component of cell-mediated acquired immunity and play an import-ant role in the defense against a number of viralinfections. Both of the T-cell subsets, T-helper (Th;CD4+) and T-cytotoxic (Tc; CD8+), first increase andthen decrease after severe, acute exercise (Steensburget al. 2001). The Th cells provide a series of cytokinesthat are essential to regulate the immune response.Tc cells kill virally infected or tumor cells by directcontact. Each of these T-cell subsets are furtherclassified by the cytokines they produce. A charac-teristic signature of a type 1 cytokine response, isproduction of interferon-γ (IFN-γ) and interleukin-2(IL-2), while type 2 cytokines include IL-4 and IL-6. It is the balance between these sets of T-cells and their cytokines that influence whether theimmune response will be more directed to either a cell-mediated (type 1) or a humoral or antibody-

Page 393: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 377

mediated (type 2) response. It is this balance that isessential for assuring an effective immune defense.It has been reported (Steensburg et al. 2001) that the Th1 cells decrease while the Th2 are largelyunaffected after acute exercise stress. Further-more, Ibfelt et al. (2002) found that the decrease may be due largely to a decrease in memory cells(CD45RO+). Such a decrease in both memory cellsand the type 1 cytokines associated with an exercise-stressed immune system might result in the inabil-ity of one to mount an effective response against aviral infection.

lymphocyte activation and apoptosis

in culture

Upon recognition of a specific antigen in vivo, both T- and B-lymphocytes become activated, grow intolarge blast cells and undergo several rounds of celldivision. This clonal expansion of antigen-specificlymphocytes assures that the cell numbers will beadequate for mounting an effective defense. Thisclonal expansion is mimicked in vitro by the stimula-tion of lymphocytes with polyclonal mitogens. The amount of DNA synthesis carried out by thesestimulated cells has long been used as an index of invivo cell-mediated immune function. Based on thisassay, it has often been observed that intense or pro-longed exercise leads to a suppression of the DNAsynthesis of lymphocytes sampled from the bloodimmediately after exercise (reviewed by Mackinnon2000a). This suppression is usually short-lived andcells from blood samples taken a few hours after theend of the exercise respond normally. This kineticanalysis suggests that the observed suppressionmay be, in part, explained by the redistribution ofcells in the blood as monitored by the phenotypeanalysis. Furthermore, the measured levels of 3H-thymidine incorporation by cells (a measure ofDNA synthesis) is an average of all the cells in theculture, yet some of the lymphocytes may be non-responders or, in fact, undergo cell death.

Research based on the use of new techniques suggests that lymphocyte cell death via apoptosismay contribute, in part, to the apparent decrease in proliferation associated with exercise (Green &Rowbottom 2003). Apoptosis is a normal and non-

inflammatory mechanism for the removal of cells ofall types within the body. For example, newly dif-ferentiated lymphocytes undergo apoptosis if theydo not encounter their cognate antigen. In a recentpublication, Green & Rowbottom (2003) suggestedthat increased apoptosis may account for an appar-ent decrease in cell proliferation. By combining vitalstaining with a fluorescent dye (CFSE) and flowcytometric analyses, this group found that exercisedid not affect cell division but rather increased celldeath. Thus, in this study, the overall observeddecrease in lymphocyte expansion was interpretedto be the net result of cell division and apoptosis,with apoptosis being the greatest contributor. This study was limited in the number of samplesanalyzed so it remains to be seen if the finding isuniversal.

In another case in which apoptosis was directlyexamined with the use of annexin V, a proteinwhich binds to the membrane of apoptotic cells,exhaustive but not moderate exercise increasedlymphocyte apoptosis (Mooren et al. 2002). Thereare various reports that exhaustive exercise bringsabout cellular changes which may participate in theapoptotic process such as DNA damage, increasedCa2+ levels (Mooren et al. 2001), ROS, cortisol anddeath–death ligand molecules. However, which oneof these is cause of apoptosis and which is the effectof apoptosis remain to be determined. Whether the observed apoptosis is indicative of a state ofimmunosuppression or is simply a normal regulat-ory mechanism associated with exercise-inducedstress, is not known.

natural killer cells

NK cells, which make up about 5–10% of the circu-lating white blood cell population, offer an immedi-ate and major response to the presence of virallyinfected cells and are believed to offer surveillanceagainst cancer. Both NK-cell number and activityhave been examined in numerous acute and chronicexercise studies. As is described below, some studieshave described major effects of exercise on NK cellswhile others do not. Faced with the plethora ofinformation regarding NK cells and their responseto exercise, Shephard & Shek (2000b) recently

Page 394: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

378 chapter 26

carried out a meta-analysis of the existing literature.After considering many parameters, including thetype of exercise, they concluded that sustained,moderate exercise usually causes an increase in cir-culating NK cells during exercise, followed by adecrease within about 1 h, and recovery to normallevels within 2 h of the end of the exercise. Both sustained vigorous exercise and a short bout of vigorous exercise brought about the same pattern of changes in NK-cell numbers, but the magnitudeof these changes were greater with the short bout ofmoderate exercise, with no apparent effect of train-ing on these numbers. These changes in circulatingNK-cell numbers have been attributed to changes incardiac output and to increased synthesis of cate-cholamines associated with the exercise.

There also are reports of increases in NK-cellcytolytic activity as well as increases in their numbers (Mackinnon 2000a). Does this change inactivity reflect only the change in cell number ordoes activity on a per cell basis actually change? For most, but not all studies, it appears that, on a percell basis, the activity is not affected (Miles et al.2002). In summary, an acute bout of exercise leads to a transitory increase and then suppression of NK-cell counts and associated cytolytic activity.However, there is no evidence that these fluctua-tions in number and function affects overall health.Authors of the meta-analysis study recommendsampling during exercise and beyond the 24-hrecovery period and to measurements of cate-cholamines, cortisol and prostaglandins in order to more definitively examine how exercise affectsNK cells.

humoral immunity immunoglobulin

levels

The humoral immune response is mediated by B-lymphocytes, the immune cells of the body that pro-duce immunoglobulins more commonly referred to as antibodies. These antibodies are glycoproteinmolecules that function as very specific cell mem-brane receptors for foreign molecules (antigens)that are encoded for by all pathogens and, in somecases, may be present on tumor cells. Upon bind-ing of the antibody to the cell-associated antigenic

receptor, the B-lymphocyte becomes activated anddevelops into a plasma cell that, in turn, secreteslarge quantities of antibodies into the blood, saliva,and other mucosal tissues. These antibodies canprotect the host in several ways including neutraliz-ing viruses, targeting bacteria for opsonization byphagocytes, activating complement, and binding toand clearing foreign particles. The immunoglobulinG (IgG) class of antibody makes up the largest proportion of serum immunoglobulins and plays a critical role in each of the above mechanisms ofprotection.

In general, the circulating levels of IgG or total immunoglobulins have been found not tochange following acute exercise or chronic training(Mackinnon 2000a). Although there is a report oflow serum immunoglobulin levels in elite swim-mers (Gleeson et al. 1995), the production of anti-bodies in response to vaccination is not suppressedand there is no other obvious immunosuppressionin these individuals (Gleeson et al. 1996; Bruunsgaardet al. 1997b). Thus, exercise does not seem to affectthe overall B-cell-mediated component of the im-mune response.

mucosal immunity

Immunoglobulins of the IgA and IgM subtypes areenriched in mucosal tissues such as the linings of theoral and gastrointestinal cavities. These immuno-globulins provide an important role in the protec-tion from pathogens that enter the body with bothair and food. In contrast to total serum immuno-globulin, salivary IgA levels have been found to bereduced after an acute bout of exercise in high per-formance athletes (reviewed by Gleeson & Pyne2000). This observation was first published in 1982,by Tomasi et al. (1982) who reported decreasedsecretory IgA in cross-country skiers. The originalthought of these investigators was that part of thisdecrease was due to cold temperatures. However,reduced levels of salivary IgA have since been measured in other athletes with intensive trainingschedules such as elite swimmers, tennis players,marathon runners, rowers, cyclists, hockey playersand kayakers (reviewed by Pedersen & Hoffman-Goetz 2000). Generally, the levels return to pre-

Page 395: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 379

exercise levels within about an hour, but in somecases, chronically depressed levels of salivary IgAwere associated with long-term training. Moderateexercise does not appear to bring about thesechanges. Salivary IgM levels mirrored the changesin IgA. Salivary levels of IgA are of special interestbecause of the high incidence of URTIs in athletesinvolved in high intensity training. Yet, it has notbeen easy to directly link reduced salivary IgA withincreased risk of URTI (Pyne et al. 2001; Novas et al.2003). There are only a few studies that indicate ageneral risk associated with exercise and suggestthat salivary IgA and IgM can be used to predictinfection. The mechanisms by which reduced IgA isrelated to infection are speculative. The lower levelsof salivary IgA may reflect secretion rates and fluidchanges brought about by the exercise. These lowerlevels did not appear to be linked to increases in cortisol caused by the exercise (McDowell et al. 1992;Dimitriou et al. 2002). Other stress-induced mole-cules such as prostaglandins may be involved(Tvede et al. 1989).

Athlete versus non-athlete

If exercise affects the immune system, is theimmune system of a well-trained athlete differentthan a non-athlete? The simple answer is ‘no’. By thevarious criteria used to assess immunity, no majordifferences have been identified. The adaptiveimmune system remains unaffected for the mostpart (Nieman & Pedersen 1999). Although a heavybout of acute exercise even in the trained athletecauses transient changes in immune cells in theblood, there is no compelling evidence that thesechanges are lasting or affect immune system func-tion. Furthermore, although it has been suggestedthat this stress-like response leaves the athlete witha somewhat suppressed immune system, vaccina-tion, which involves the co-ordinated function ofboth B- and T-lymphocytes, has been shown not tobe affected in male triathletes (Bruunsgaard et al.1997b). On the other hand, a skin test (delayed typehypersensitivity [DTH]) response to recall antigenwas shown to be decreased when carried out imme-diately after completing a half-ironman triathloncompetition (Nieman & Pedersen 1999). To inter-

pret these findings it is necessary to remember thatthe DTH response is a short-term response while an immune response to vaccination involves a much longer period. Furthermore, another studycarried out in normal, healthy individuals, indicatedthat high intensity progressive resistance trainingdid not affect a DTH response (Rall et al. 1996).

The idea that training may be immunosuppress-ive is based on the observation that overtraining or exhaustive training sometimes encountered bymarathon runners or long-distance swimmers isoften associated with URTIs. These athletes alsomay show decreased levels of salivary IgA and IgM.Thus, it is concluded that the infection is due to the reduction in the salivary immunoglobulins (seeMackinnon 2000b). In a recent review, Smith (2003)proposed that the URTIs associated with over-training or excessive exercise, might be associatedmore with tissue trauma and the production ofstress molecules and hormones than with reducedimmunoglobulin levels. Smith suggests that thetraining brings about type 2 (Th2) cytokine produc-tion and suppresses the production of type 1 (Th1) -associated cytokines. This balance of cytokineswould put the immune system in a compromisedposition for fighting off viral infections. While thishypothesis is supported by data in the literature,there is still the need to demonstrate that cytokineresponses are affected by overtraining and thatURTIs are the outcome. In summary, the majority ofinformation suggests that the immune system of thetrained athlete is not substantially different fromthat of a healthy sedentary person. However, excess-ive or overtraining may, on a short-term basis, make the athlete more susceptible to URTIs but notnecessarily due to immunosupression (Weidner et al. 1998).

Mechanisms for exercise–immune interactions—the role of cytokines

Although there is a variety of observations thatpoint to exercise as a regulator of immune func-tion, the molecular mechanisms that underlie thisinteraction of exercise and immunity remain to befully elucidated. On the other hand, there is evid-ence that products of the neuroendocrine system

Page 396: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

380 chapter 26

that are elicited by exercise can affect immune function.

It has long been recognized that leukocyte-derived cytokines act as major communicators andregulators of the immune system (reviewed byVilcek 2003). These low molecular weight proteinsor glycoproteins secreted by many cells in the bodyin response to various stimuli are present in lowconcentrations in the blood. In contrast to the classicendocrine-derived hormones which act on their target cells and tissues at distant sites, cytokinestypically act locally in either an autocrine or para-crine manner. Many of these cytokines retain thename ‘interleukin’ (e.g. interleukin-2 or IL-2), thusimplying their production by a leukocyte to theirability to act on other leukocytes. Because they canact pleitropically and may be redundant in function,it is often difficult to pinpoint their exact mechan-ism of action. In fact, we now know that cytokinesproduced by immune cells not only provide com-munication among cells of the immune system but also serve as a means of communication amongthe immune, nervous, and endocrine systems. Theimpact of exercise on cytokine production explains,in part, how exercise can modulate both immuneand neuroendocrine systems.

Initially, assays of biological activity (bioassays)were used to identify cytokines and growth factorsin the plasma or serum following exercise. In 1983,researchers (Cannon & Kluger 1983) showed thatthe plasma obtained from subjects following exer-cise, when injected into rats, caused an increase inbody temperature. This bioassay for cytokines thatproduce fever, indicated the presence of IL-1 or a related cytokine Both the development of moresensitive assays for this and other cytokines and theproduction of recombinant molecules has suggestedroles for many other cytokines following exercise(Pedersen & Toft 2000). Furthermore the plethora ofobserved exercise-induced increases in cytokinesled many to suggest that exercise causes an in-flammatory-like response. One such inflammation-related cytokine that is produced in large amountsafter exercise, and thus has been the subject of manystudies, is IL-6 (Pedersen & Toft 2000). Although IL-6 has been categorized as a proinflammatory mole-cule it also has many anti-inflammatory properties

such as the ability to inhibit tumor necrosis factor-α(TNF-α) and IL-1 and the induction of IL-1 receptorantagonist. At one point, it was believed that exer-cise-induced IL-6 resulted from muscle damage(Bruunsgaard et al. 1997a; Pedersen 2000). Whilethis may indeed be true, muscle damage is not necessary to observe increases in IL-6. Although acorrelation has been reported between the exercise-induced increases in both IL-6 and epinephrine, the infusion of epinephrine did not mimic the extentor the kinetics of changes in IL-6 found with exer-cise (Steensberg et al. 2001). IL-6 is one of manycytokines (e.g. IL-1, TNF-α, IFN-γ, IL-12) that areknown to stimulate the HPA axis. As is outlinedabove, the products of the HPA axis, in particularthe glucocorticoids, are able to have multiple effectson immune function. IL-6 has a direct link to theHPA axis given that pituitary cells express receptorsfor IL-6 and respond to its presence (Besedovsky &Del Rey 2001). Receptors for IL-6 and others cyto-kines have been identified in the brain as well.Interestingly, Nybo et al. (2002), found that the brain itself could release IL-6 following a bout ofprolonged exercise, albeit at a much lower level(approximately 100-fold less) than skeletal muscle.

In a recent review article, Suzuki et al. (2002) summarized the results of a number of studies inwhich circulating cytokine levels were detected inthe plasma following various exercise regimens.However, a major consideration presented in thisreview was whether or not measuring the plasmalevels of cytokines provides sufficient informationsince both the localization and utilization of cyto-kines in specific organs and tissues (both immuneand non-immune) are also important in assessingcytokine contribution to overall immune function.Regardless, in approximately one-half of these studies, the proinflammatory molecule TNF-α wasshown to increase. Plasma IL-6 concentrations alsoincreased with the greatest increases usually occur-ring with exercise that caused potential muscledamage. The anti-inflammatory cytokines inter-leukin-1 receptor antagonist (IL-1ra) and IL-10 wereseen to increase in many studies. In contrast, none ofthese studies reported a change in IFN-γ; some evenfound a decrease in IL-2.

In order to try to reconcile some of these conflict-

Page 397: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 381

ing reports, these same investigators (Suziki et al.2002) selected 16 target cytokines, including pro-and anti-inflammatory, immunomodulatory, multi-functional, colony stimulating and chemokines.They measured both plasma and urine concentra-tions at 10 min and 2 and 24 h following a short-duration maximal exercise. The findings weresurprising. For example, although plasma TNF-αwas not detected, the levels of TNF-α in the urineincreased fivefold. An opposite finding was seenwith IL-1β, where concentrations increased in theplasma but were undetectable in the urine. IL-2, IL-12, IFN-α and IFN-γ either were not detected or did not change in either plasma or urine. The anti-inflammatory cytokines changed more dramaticallyin response to exercise. For example, IL-1ra increasedin plasma and urine as did IL-4. IL-6 increased inboth fluids but only significantly in urine. While the proinflammatory cytokines TNF-α and IL-1increased, they showed a delayed response com-pared with anti-inflammatory cytokines, particu-larly IL-1ra. Together, these results indicate thatbecause of the difference in kinetics of production,half-life and clearance, sampling one cytokine at atime either during or after exercise is likely to pro-vide misleading results.

The impact of exercise on diseases that areimmunologically resisted

The immune system has long been known to play akey role in the resistance to a variety of infectiouspathogens. Although not as well-defined, there isalso evidence of a role for immune function in thesuccessful defense against cancer. However, oftenforgotten are the undesirable contribution of theimmune system in conditions such as allergy andautoimmunity. Thus, the impact of exercise onimmune function can range from beneficial to detrimental depending on the nature of the exerciseand the role of the immune system in the disease ofinterest. For many years, there was only anecdotalevidence that exercise can affect the developmentand/or progression of diseases that have an im-munological component. However, only recently hasthere been experimental proof that such a relation-ship does indeed exist.

viral infections

The influence of the neuroendocrine system onimmunity to infectious pathogens has been betterdefined during the past decade. Many of the studiesto define the mechanisms underlying this relation-ship have relied on the use of stress-based models in mice (Sheridan et al. 1998; Bonneau et al. 2001).Although far fewer studies have explored theimpact of stress on the development of infections inhumans (Cohen & Herbert 1996; Cohen & Miller2001), the significance of these studies is broad.Stress and other psychosocial factors have also beenshown to influence the ability of vaccinations forhepatitis B virus and influenza virus to elicit vaccine-specific immune responses (Yang & Glaser2002).

Although there is clearly a role for stress–neuroendocine–immune interactions in the defenseagainst infectious disease, there is little evidence inhumans that moderate exercise plays a signific-ant role in the defense against viral and bacterialinfections. For example, a recent study (Weidner & Schurr 2003) found that normally sedentary buthealthy individuals did not exhibit differences inthe type, severity, of duration of cold symptomswith the addition of exercise. Human immuno-deficiency virus (HIV) infection is directly related tothe immune system since HIV infects immune cells,i.e. T-cells and macrophages. While exercise mayhelp boost the CD4 levels of HIV+ individuals itdoes not reverse the disease. Likewise, exercise canincrease the general well-being of individuals at allages but does not reverse or prevent age or diseaseinfluenced changes.

There have also been a number of recent murine-based studies conducted in an attempt to define the impact of exercise on the immune response toand susceptibility to viral pathogens. Although thefindings of these studies vary depending on the type of exercise and the nature of the pathogen, theyhave been valuable in dissecting the many effects of exercise on immune-related processes includ-ing cytokine (Davis et al. 1998; Kohut et al. 1998,2001a, 2001b) and antibody (Kohut et al. 2001a) pro-duction, antigen presentation (Ceddia & Woods1999), macrophage function (Davis et al. 1997) and

Page 398: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

382 chapter 26

macrophage chemotaxis (Ortega et al. 1997). Suchanimal-based studies will continue to provide awealth of information in helping to define the cel-lular and molecular mechanisms underlying theinteractions among exercise, immunity and suscept-ibility to infectious pathogens.

exercise and cancer

Does exercise reduce the risk of cancer? The simpleanswer is ‘yes’. Many, but not all, epidemiologicalstudies have indicated that there is indeed a correla-tion between physical activity and a reduced risk of cancer (reviewed by Shephard & Shek 1995,2000a; Gammon et al. 1998; McTiernan et al. 2003). Is this correlation due to a direct affect of exercise on the immune system and its ability to prevent thedevelopment and/or progression of cancer? Whileit is possible, there is no convincing evidence forsuch a direct connection.

Several epidemiological studies suggest thatphysical activity correlates with the incidence ofvarious cancers including those of the colon, breast,prostate, testes and lung (reviewed by Gammon et al. 1998). However, not all of these studies werecarried out in the same way. For example, somestudies included activities related to a job or dailyliving (e.g. lifting boxes or housework) whereasother studies limited assessment to leisure timeactivities (e.g. weight training, aerobic exercise).Other variables included the nature of the exercise(i.e. strenuous or moderate) and the time of life during which the activity was performed (reviewedby Gammon et al. 1998). A recent study (McTiernanet al. 2003), to determine the impact of strenuous ormoderate recreational physical activity on the riskof breast cancer, indicated that risk decreased withexercise of longer duration and that the exercise didnot have to be strenuous. This study was carried outwith post-menopausal women but other studieshave considered exercise at various ages of life(reviewed by Gammon et al. 1998). Whereas some of these studies found a correlation with age (e.g.Thune et al. 1997) others did not (e.g. Verloop et al.2000). Age is an especially important considerationfor women because age inherently brings about achange in reproductive hormones, a confounding

variable for several types of cancer. A reduction incirculating hormones and disruption of the men-strual cycle in some women undergoing strenuousexercise has been invoked as a mechanism by whichexercise can directly lessen the risk of breast cancer(reviewed by Hoffman-Goetz et al. 1998). However,few women exercise to this extent. Another recentstudy with women who carry mutations in BRCA1or BRCA2 genes, and thus putting them at a higherrisk for cancer, indicated that they may be protectedfrom or delay the onset of breast cancer by remain-ing active and maintaining a healthful weight whileyoung (King et al. 2003).

Depending on the intensity, exercise may alsobring about increased levels of catecholamines, glucocorticoids, β-endorphins, growth hormoneand prolactin in the circulation. These hormones allhave a direct link to cells of the neuroendocrine andimmune systems but the link to cancer is much moretenuous. Likewise, the changes in the levels of NKcells or other leukocytes in the circulating bloodcaused by intense exercise have never been directlylinked to susceptibility to any disease, let alone cancer. It should be noted that epidemiologicalstudies are confounded by diet, body build, healthhistory and other countless factors that can con-tribute to the development and progression of cancer. Thus, there is no simple answer to explainhow an active life style may protect against cancera

if it even does!

autoimmunity

If exercise can modulate immune responsivenesscan it affect autoimmunity? There are many dis-eases with an underlying autoimmune pathology;for example, diabetes, multiple sclerosis, rheuma-toid arthritis. Some of these can be reproduced inanimal models. There is anecdotal and experimentalevidence to correlate some aspects of exercise stresswith autoimmunity, but in general, exercise did not affect the development or cause of autoimmunediseases (Ferry 1996). The impact of stress and theneuroendocrine system on aspects of autoimmunityhas been well documented (Ligier & Sternberg 2001;Prat & Antel 2001; Rogers & Brooks 2001; Wilder &Elenkov 2001). Therefore, it would be expected that

Page 399: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 383

exercise, though one or more components of theneuroendocrine system, is able to regulate both thedevelopment and progression of autoimmune dis-ease. Exactly how this regulation takes place andhow exercise can be used to control this regulationremains to be determined.

The impact of exercise on other systemsthat may indirectly affect the immunesystem

The body is clearly a well-integrated set of systems(Fig. 26.3). It would be unreasonable to assume thatall effects of exercise on immunity are mediatedthrough neuroendocrine pathways. For one, themuscular system is obviously a prominent target ofexercise. During exercise, the muscles compete withother systems of the body for energy and also pro-duce waste. It has been suggested that the depletionof glycogen, glucose and glutamine from musclemakes these compounds less available to the con-tinually renewing cells of the immune system.Moreover, waste products such as lactate may neg-atively affect lymphocyte metabolism (Miles et al.2003). Muscle damage, likely to occur during certainkinds of intensive exercise, also initiate the synthesisof inflammatory cytokines. Furthermore, the releaseof heat shock proteins by exercise stressed cells may act as ‘danger signals’ for the immune system(Moseley 2000) and activate antigen-presentingcells and lymphocytes. The cardiovascular system isresponsible for transporting oxygen, nutrients andwastes, and responds very rapidly to exercise. Heartrate and blood pressure increase to assist the flow ofblood. As exercise causes fluids to leave the major

vascular compartments, the blood and plasma vol-umes decrease. This hemoconcentration causes anincrease in hemoglobin and cells in the blood.Furthermore, blood may be directed away fromperipheral immune organs like the spleen with thenet effect that the numbers of leukocytes in theblood is increased (reviewed by Nielsen 2003).

Just as the heart rate rapidly increases, ventilationalso rapidly rises and is proportional to changes inintensity in the exercise. The trachea enlarges toallow more, unrestricted air flow. This increased air flow may dry the mouth and have deleteriouseffects on the lining of the trachea, as well as limitthe amount of salivary immunoglobulins. Thesechanges may partially explain why upper respirat-ory infections increase with intensive, long endur-ance exercise.

Nielsen (2003) nicely discusses the lymphocyteresponse to maximal exercise from the point of viewof the body’s cardiopulmonary responses. He sug-gests that the transient changes in cell numbers fol-lowing acute exercise can be explained by increasedblood flow, and that increases in ventilation andblood pressure may affect the respiratory epithe-lium making it more susceptible to infection.

Summary and conclusions

There is no doubt that exercise impacts the neuro-endocrine–immune axis. Exactly how, where and to what extent exercise acts on this complex inter-related system, remains to be further defined.Although there is a plethora of indicators ofimmunomodulation, the ultimate question is howthese indicators are related to the functioning of theimmune system and how this functioning is relatedto overall health. Correlations between exercise andchanges in immune parameters do not necessarilytranslate to cause and effect. Even if they did, westill might not understand when it is good to activ-ate or to dampen an immune function, particularlyan inflammatory response.

It is difficult enough to evaluate the general statusof the immune system in a healthy, homeostatic condition, let alone under stress from exercise. Weare able recognize a ‘broken’ immune system whenwe see one, for example an immune system ravaged

Neuroendocrine-derivedhormones

Cytokines

Musclemetabolism

Cardiopulmonary/hemodynamics

Exercise Immunesystem

Fig. 26.3 Mechanisms of exercise-mediated modulation ofthe immune system.

Page 400: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

384 chapter 26

by the HIV, inherited or acquired immunodeficien-cies and autoimmune disorders. In contrast, we donot have a good handle on how to evaluate anapparently intact system. Illness is not necessarilyan indicator of a failed immune system. Instead, itmay well indicate that the immune system is appro-priately responding to fight the acute phase of aninfectious disease and is establishing immunolo-gical memory in preparation for when the host ischallenged with the infectious pathogen once again.

For obvious ethical considerations, we can neitherchallenge humans with a pathogen nor extensivelysample immune compartments where immune res-ponses to such pathogens generally occur. In theserespects, studies in animals are valuable in that they

provide a means by which to model the impact ofexercise on immunity. In addition, newer techniquesand an expanding knowledge of the immune sys-tem have allowed scientists in the field to begin tounravel some of the mysteries underlying immunefunction. It will take insightful collaborations be-tween scientists from the disciplines of kinesiology,immunology and neuroendocrinology to move thefield forward. The goal is to reach the point wherethe prescription is no longer a general ‘get plenty ofexercise’, but one in which a very specific recom-mendation is given of the intensity, mode, durationand kind of exercise needed to complement a stand-ard regimen of medical intervention in an attemptenhance immune function.

References

Ader, R. & Cohen, N. (1975) Behaviorallyconditioned immunosuppression.Psychosomatic Medicine 37, 333–340.

Ader, R., Felten, D.L. & Cohen, N. (eds.)(2001) Psychoneuroimmunology, 3rd edn.Academic Press, San Diego, CA.

Bellinger, D.L., Lorton D., Felten S.Y. & Felten, D.L. (1992) Innervation oflymphoid organs and implications indevelopment, aging, and autoimmunity.International Journal ofImmunopharmacology 14, 329–344.

Ben-Eliyahu, S. & Shakhar, G. (2001) Theimpact of stress, catecholamines, and the menstrual cycle on NK activity andtumor development: from in vitrostudies to biological significance. In:Psychoneuroimmunology, 3rd edn. (Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA: 545–563.

Berczi, I., Chow, D.A., Baral, E. & Nagy E.(1998) Neuroimmunoregulation andcancer. International Journal of Oncology13, 1049–1060.

Bertrand, A. (2003) ‘Healing power,exercise helps relieve fatigue and othersymptoms of disease’. St. Louis PostDispatch reported in Centre Daily Times.Monday, September 15, 2003.

Besedovsky, H.O. & Del Rey, A. (2001)Cytokines as mediators of central andperipheral immune-neuroendocrineinteractions. In: Psychoneuroimmunology,3rd edn. (Ader, R., Felten, D.L. & Cohen,N., eds.). Academic Press, San Diego,CA: 1–17.

Blalock, J.E. (1994). Shared ligands and

receptors as a molecular mechanism forcommunication between the immuneand neuroendocrine systems. Annals ofthe New York Academy of Sciences 741,292–298.

Bonneau, R.H., Padgett, D.A. & Sheridan,J.F. (2001) Psychoneuroimmuneinteractions in infectious disease: studiesin animals. In: Psychoneuroimmunology,3rd edn. (Ader, R., Felten, D.L. & Cohen,N., eds.). Academic Press, San Diego,CA: 483–497.

Booher, M.A. & Smith, B.W. (2003)Physiological effects of exercise on thecardiopulmonary system. Clinics inSports Medicine 22, 1–21.

Bruunsgaard, H., Galbo, H., Halljaer-Kristensen, J. et al. (1997a) Exercise-induced increases in serum interleukin 6 in humans is related to muscledamage. Journal of Physiology 499,833–841.

Bruunsgaard, H., Hartkopp, A., Mohr, T. et al. (1997b) In vivo cell mediatedimmunity and vaccination responsefollowing prolonged intense exercise.Medicine in Science and Sports and Exercise29, 1176–1181.

Buckingham, J.C., Gillies, G.E. & Cowell, A.-M. (eds.) (1997) Stress, Stress Hormones, and the Immune System.John Wiley & Sons, New York.

Cannon, J.G. & Kluger, M.J. (1983)Endogenous pyrogen activity in humanplasma after exercise. Science 220,617–619.

Ceddia, M.A. & Woods, J.A. (1999)Exercise suppresses macrophage

antigen presentation. Journal of AppliedPhysiology 87, 2253–2258.

Chambers, D.A. & Schauenstein, K. (2000) Mindful immunology:neuroimmunomodulation. Trends inImmunology 21, 168–169.

Chambers, D.A., Cohen, R.L. & Perlman,R.L. (1993) Neuroimmune modulation:signal transduction and catecholamines.Neurochemistry International 22, 95–110.

Chrousos, G.P. & Gold, P.W. (1992) Theconcept of stress and stress systemsdisorders; overview of physical andbehavioral homeostasis. Journal of theAmerican Medical Association 267,1244–1252.

Cohen, S. & Herbert, T.B. (1996) Healthpsychology: psychological factors andphysical disease from the perspective of human psychoneuroimmunology.Annual Review of Psychology 47, 113–142.

Cohen, S. & Miller, G.E. (2001) Stress,immunity, and susceptibility to upper respiratory infection In:Psychoneuroimmunology, 3rd edn. (Ader,R., Felten, D.L. & Cohen, N., eds.).Academic Press, San Diego, CA:499–509.

Conti, A., Maestroni, G.J.M., McCann, S.M.et al. (eds.) (2000) Neuroimmunology:Perspectives at the New Millennium, Vol. 917. The New York Academy ofSciences, New York.

Davis, J.M., Kohut, M.L., Colbert, L.H. et al.(1997) Exercise, alveolar macrophagefunction, and susceptibility torespiratory infection. Journal of AppliedPhysiology 83, 1461–1466.

Page 401: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 385

Davis, J.M., Weaver, J.A., Kohut, M.L. et al.(1998) Immune system activation andfatigue during treadmill running: role of interferon. Medicine and Science inSports and Exercise 30, 863–868.

De Kloet, E.R., Oitzl, M.S. & Schobitz, B.(1994) Cytokines and the braincorticosteroid receptor balance:relevance to pathophysiology of neuroendocrine–immunecommunication.Psychoneuroendocrinology 19, 121–134.

Dhabhar, F.S. & McEwen, B.S. (2001)Bidirectional effects of stress andglucocorticoid hormones on immunefunction. In: Psychoneuroimmunology, 3rd edn. (Ader, R., Felten, D.L. & Cohen,N., eds.). Academic Press, San Diego,CA: 301–338.

Dimitriou, L., Sharp, N.C. & Dougherty,M. (2002) Circadian effects on the acuteresponses of salivary cortisol and IgA inwell trained swimmers. British Journal ofSports Medicine 36, 260–264.

Felten, D.L. & Felten, S.Y. (1988)Sympathetic noradrenergic innervationof immune organs. Brain, Behavior, andImmunity 2, 293–300.

Felten, D.L., Felten, S.Y., Bellinger, D.L. & Madden, K.S. (1993) Fundamentalaspects of neural–immune signaling.Psychotherapy and Psychosomatics 60,46–56.

Ferry, A. (1996) Exercise and autoimmuinediseases. In: Exercise and ImmuneFunction (Hoffman-Goetz, L., ed.). CRC Press, Boca Raton, FL: 163–178.

Fox, K.R. (1999) The influence of physicalactivity on mental well-being. PublicHealth Nutrition 2, 411–418.

Gabriel, H., Schmitt, B., Urhausen, A. & Kindermann, W. (1993) IncreasedCD45RA+ CD45RO+ cells indicateactivated T cells after enduranceexercise. Medicine and Science in Sportsand Exercise 25, 1352–1357.

Gammon, M.D., John, E.M. & Britton, J.A.(1998) Recreational and occupationalphysical activities and risk of breastcancer. Journal of the National CancerInstitute 90, 100–117.

Garza, H.H. & Carr, D.J. (1997)Neuroendocrine peptide receptors oncells of the immune system. ChemicalImmunology 69, 132–54.

Glaser, R. & Kiecolt-Glaser, J. (eds.) (1994) Handbook of Human Stress andImmunity, Academic Press, San Diego,CA.

Gleeson, M. & Pyne, D.B. (2000) Specialfeature for the Olympics: effects ofexercise on the immune system:

American Journal of Physiology 275,L1089–L1094.

Kohut, M.L., Boehm, G.W. & Moynihan,J.A. (2001a) Moderate exercise isassociated with enhanced antigen-specific cytokine, but not IgM antibodyproduction in aged mice. Mechanisms of Ageing and Development 122,1135–1150.

Kohut, M.L., Boehm, G.W. & Moynihan,J.A. (2001b) Prolonged exercisesuppresses antigen-specific cytokineresponse to upper respiratory infection.Journal of Applied Physiology 90, 678–684.

Lakier-Smith, L. (2003) Overtraining,excessive exercise, and alteredimmunity: is this a T helper-1 versus Thelper-2 lymphocyte response? SportsMedicine 33, 347–364.

Ligier, S. & Sternberg, E.M. (2001) The neuroendocrine system andrheumatoid arthritis: focus on thehypothalamo–pituitary–adrenal axis. In: Psychoneuroimmunology, 3rd edn. (Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA: 449–469.

Livnat, S., Felten, S.Y., Carlson, S.L.,Bellinger, D.L. & Felten, D.L. (1985)Involvement of peripheral and central catecholamine systems inneural–immune interactions. Journal ofNeuroimmunology 10, 5–30.

McDowell, S.L., Hughes, R.A., Hughes,R.J. et al. (1992) The effect of exhaustiveexercise on salivary immunoglobulin A.Journal of Sports Medicine and PhysicalFitness 32, 412–415.

Mackinnon, L.T. (2000a) Exerciseimmunology: current issues. In:Nutrition and Exercise Immunology(Nieman, D.C. & Pedersen, B.K., eds.).CRC Press, Boca Raton, FL: 3–24.

Mackinnon, L.T. (2000b) Overtrainingeffects on immunity and performance inathletes. Immunology and Cell Biology 78,502–509.

McTiernan, A., Kooperberg, C., White, E. et al. (2003) Recreational physical activity and the risk of breast cancer inpostmenopausal women. Journal of theAmerican Medical Association 290,1331–1336.

Madden, K.S., Sanders, V.M. & Felten, D.L.(1995) Catecholamine influences andsympathetic neural modulation ofimmune responsiveness. Annual Reviewof Pharmacology and Toxicology 35,417–448.

Marsland, A.L., Bachen E.A., Cohen, S.,Rabin, B. & Manuck, S.B. (2002) Stress,immune reactivity and susceptibility

exercise effects on mucosal immunity.Immunology and Cell Biology 78, 536–544.

Gleeson, M., McDonald, W.A., Cripps,A.W. et al. (1995) Exercise, stress andmucosal immunity in elite swimmers.Advances in Experimental Medical Biology371(A), 571–574.

Gleeson, M., Pyne, D.B., McDonald, W.A.et al. (1996) Pneumococcal antibodyresponse in elite swimmers. ClinicalExperimental Immunology 105, 238–244.

Glenister, D. (1996) Exercise and mentalhealth: a review. Journal of the RoyalSociety of Health 116, 7–13.

Green, K.J. & Rowbottom, D.G. (2003)Exercise-induced changes to in vitro T-lymphocyte mitogen responses usingCFSE. Journal of Applied Physiology 95,57–63.

Greer, S. (2000) What’s in a name?Neuroimmunomodulation orpsychoneuroimmunology. In:Neuroimmunology: Perspectives at the New Millennium, vol. 917 (Conti, A.,Maestroni, G.J.M., McCann, S.M. et al., eds.). The New York Academy of Sciences, New York: 568–574.

Hoffman-Goetz, L. (ed.) (1996) Exercise and Immune Function. CRC Press, BocaRaton, FL.

Hoffman-Goetz, L. & Pedersen, B.K. (1994) Exercise and the immune system: a model of the stress response?Immunology Today 15, 382–387.

Hoffman-Goetz, L. & Pedersen, B.K. (2001)Immune responses to acute exercise:hemodynamic, hormonal and cytokineinfluences In: Psychoneuroimmunology,3rd edn. (Ader, R., Felten, D.L. & Cohen,N., eds.). Academic Press, San Diego,CA: 123–132.

Hoffman-Goetz, L., Apter, D., Demark-Wahnefried, W. et al. (1998) Possiblemechanisms mediating an associationbetween physical activity and breastcancer. Cancer 83, 621–628.

Ibfelt, T., Petersen, E.W., Bruunsgaard, H.,Sandmand, M. & Pedersen, B.K. (2002)Exercise-induced change in type 1cytokine-producing CD8+ T cells isrelated to a decrease in memory T cells.Journal of Applied Physiology 93, 645–648.

King, M.D., Marks, J.H., Mandell, J.B. &New York Breast Cancer Study Group.(2003) Breast and ovarian cancer risksdue to inherited mutations in BRCA1and BRCA2. Science 302, 643–646.

Kohut, M.L., Davis, J.M., Jackson, D.A. et al. (1998) Exercise effects on IFN-βexpression and viral replication in lungmacrophages after HSV-1 infection.

Page 402: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

386 chapter 26

to infectious disease. Physiology andBehavior 77, 711–716.

Miles, M.P., Mackinnon, L.T., Grove, D.S.et al. (2002). The relationship of naturalkiller cell counts, perforin mRNA andCD2 expression to post-exercise naturalkiller cell activity in humans. ActaPhysiologica Scandinavica 174, 317–325.

Miles, M.P., Kraemer, W.J., Nindl, B.C. et al. (2003) Strength, workload,anaerobic intensity and the immuneresponse to resistance exercise inwomen. Acta Physiologica Scandinavica178, 153–163.

Mooren, F.C., Lechtermann, A., Fromme,A., Thorwestern, L. & Völker, K. (2001)Alterations in intracellular calciumsignaling of lymphocytes afterexhaustive exercise. Medicine and Sciencein Sports and Exercise 33, 242–248.

Mooren, F.C., Blöming, D., Lechtermann,A., Lerch, M.M. & Völker, K. (2002)Lymphocyte apoptosis after exhaustiveand moderate exercise. Journal of AppliedPhysiology 93, 147–153.

Moseley, P.L. (2000) Exercise, stress, andthe immune conversation. Exercise andSports Sciences Reviews 28, 128–132.

Moynihan, J.A. (2003) Mechanisms ofstress-induced modulation of immunity.Brain, Behavior and Immunity 17 (suppl.1), S11–S16.

Moynihan, J.A. & Ader, R. (1996).Psychoneuroimmunology: animalmodels of disease. PsychosomaticMedicine 58, 546–558.

Moynihan, J.A. & Stevens, S.Y. (2001)Mechanisms of stress-inducedmodulation in animals. In:Psychoneuroimmunology, 3rd edn. (Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA:227–250.

Nielsen, H.B. (2003) Lymphocyteresponses to maximal exercise: aphysiological perspective. SportsMedicine 33, 853–867.

Nieman, D.C. (2003) Current perspectiveon exercise immunology. Current SportsMedicine Reports 2, 239–242.

Nieman, D.C. & Pedersen, B.K. (1999)Exercise and immune function. SportsMedicine 27, 73–80.

Nieman, D.C. & Pedersen, B.K. (eds.)(2000) Nutrition and Exercise immunology.CRC Press, Boca Raton, FL.

Novas, A.M., Rowbottom, D.G. & Jenkins,D.G. (2003) Tennis incidence of URTIand salivary IgA. International Journal ofSports Medicine 24, 223–229.

Nybo, L., Nielsen, B., Pedersen, B.K,Møller, K. & Secher, N.H. (2002)

exercise on anxiety, depression, andsensitivity to stress: a unifying theory.Clinical Psychology Review 21, 33–61.

Sephton, S. & Spiegel, D. (2003) Circadiandisruption in cancer: a neuroendocrine–immune pathway from stress to disease?Brain, Behavior, and Immunity 17,321–328.

Shephard, R.J. & Shek, P.N. (1995) Cancer,immune function and physical activity.Canadian Journal of Applied Physiology 20,1–25.

Shephard, R.J. & Shek, P.N. (2000a) Does regular physical activity reducesusceptibility to cancer? In: Exercise andImmune Function (Shephard, R.J., ed.).Adis International, Philadelphia, PA:131–154.

Shephard, R.J. & Shek, P.N. (2000b) Effectsof exercise and training on natural killercell counts and cytolytic activity: a meta-analysis. In: Exercise and ImmuneFunction (Shephard, R.J., ed.). AdisInternational, Philadelphia, PA: 37–58.

Sheridan, J.F., Dobbs, C., Jung, J. et al.(1998) Stress-induced neuroendocrinemodulation of viral pathogenesis andimmunity. Annals of the New YorkAcademy of Sciences 840, 803–808.

Smith, L.L. (2003) Overtraining, excessiveexercise, and altered immunity. SportsMedicine 33, 347–364.

Solomon, G.F. (1969) Stress and antibodyresponse in rats. International Archives ofAllergy 35, 97–104.

Solomon, G.F. (1981a) Emotion andpersonality factors in the onset andcourse of autoimmune disease,particularly rheumatoid arthritis. In:Pyschoneuroimmunology (Ader, R., ed.).Academic Press, New York: 159–179.

Solomon, G.F. (1981b) Immunologicalabnormalities in mental illness. In:Pyschoneuroimmunology (Ader, R., ed.).Academic Press, New York: 259–278.

Solomon, G.F. & Moos, R.H. (1964)Emotions, immunity, and disease. Aspeculative theoretical integration.Archives of General Psychiatry 11,657–674.

Solomon, G.F., Moos, R.H., Fessel, W.F. & Morgan, E.E. (1966) Globulins andbehavior in schizophrenia. InternationalJournal of Neuropsychiatry 2, 20–26.

Solomon, G.F., Levine, S. & Kraft, J.K.(1968) Early experiences and immunity.Nature 220, 821–822.

Steensberg, A., Toft, A., Bruunsgaard, H. et al. (2001) Strenuous exercise decreasesthe percentage of type 1 T cells in thecirculation. Journal of Applied Physiology91, 1708–1712.

Interleukin-6 release from the humanbrain during prolonged exercise. Journalof Physiology 542, 991–995.

Ortega, E., Forner, M.A. & Barriga, C.(1997) Exercise-induced stimulation ofmurine macrophage chemotaxis: role of corticosterone and prolactin asmediators. Journal of Physiology 498,729–734.

Padgett, D.A. & Glaser, R. (2003). Howstress influences the immune response.Trends in Immunology 24, 444–448.

Paluska, S.A. & Schwenk, T.L. (2000)Physical activity and mental health:current concepts. Sports Medicine 29,167–180.

Pedersen, B.K. (2000) Exercise andcytokines. Immunology and Cell Biology78, 532–535.

Pedersen, B.K. & Hoffman-Goetz, L. (2000) Exercise and the immune system:regulation, integration and adaptation.Physiological Reviews 80, 1055–1081.

Pedersen, B.K. & Toft, A.D. (2000) Effectsof exercise on lymphocytes andcytokines. British Journal of SportsMedicine 34, 246–251.

Prat, A. & Antel, J.P. (2001)Neuroendocrine influences onautoimmune disease: multiple sclerosis.In: Psychoneuroimmunology, 3rd edn.(Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA: 433–448.

Pyne, D.B., McDonald, W.A., Gleeson, M.et al. (2001) Mucosal immunity,respiratory illness and competitiveperformance in elite swimmers. Medicineand Science in Sports and Exercise 33,348–353.

Rabin, B.S. (1999) Stress, Immune Function,and Health: the Connection. Wiley-Liss,New York.

Rall, L.C., Roubenoff, R., Canon, J.G. et al.(1996) Effects of progressive resistancetraining on immune responses in agingand chronic inflammation. Medicine andScience in Sports and Exercise 28,1356–1365.

Robergs, R.A. & Keteyian, S.J. (eds.) (2003)Fundamentals of Exercise Physiology: forFitness, Performance, and Health, 2nd edn. McGraw-Hill, Boston.

Rogers, M.P. & Brooks, E.B. (2001)Psychosocial influence. immunefunction, and the progression of autoimmune disease. In:Psychoneuroimmunology, 3rd edn. (Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA: 399–419.

Salmon, P. (2001) Effects of physical

Page 403: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise and immunity 387

Steensberg, A., Morrow, J., Toft, A.D.,Bruunsgaard, H. & Pedersen, B.K.(2002). Prolonged exercise, lymphocyteapoptosis and F2-isoprostanes. European Journal of Applied Physiology 87,38–42.

Stevens-Felten, S.Y. & Bellinger, D.L.(1997) Noradrenergic and peptidergicinnervation of lymphoid organs.Chemical Immunology 69, 99–131.

Suzuki, K., Nakaji, S., Yamoda, M. et al.(2002) Systemic inflammatory responseto exhaustive exercise. Cytokine kinetics.Exercise Immunology Review 8, 6–48.

Thune, I., Brenn, T., Lund, E. & Gaard, M.(1997) Physical activity and the risk ofbreast cancer. New England Journal ofMedicine 336(18), 1269–1275.

Tomasi, T.B., Trudeau, F.B., Czerwinski, D.& Erredg, S. (1982) Immune parametersin athletes before and after strenuousexercise. Journal of Clinical Immunology 2,173–178.

Turner-Cobbs, J.M., Sephton, S.E. &Spiegel, D. (2001) Psychological effectson immune function and diseaseprogression in cancer: Human studies.

duration of a viral upper respiratoryillness. Medicine and Science in Sports andExercise 30, 1578–1583.

Weigent, D.A., Carr, D.J. & Blalock, J.E.(1990) Bidirectional communicationbetween the neuroendocrine andimmune systems. Common hormonesand hormone receptors. Annals of theNew York Academy of Sciences 579, 17–27.

Wilder, R.L. & Elenkov, I.J. (2001) Ovarianand sympathoadrenal hormones,pregnancy, and autoimmune disease. In: Psychoneuroimmunology, 3rd edn.(Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA:421–431.

Woods, J.A., Lowder, T.W. & Keylock, K.T.(2002) Can exercise training improveimmune function in the aged? Annals ofthe New York Academy of Sciences 959,117–127.

Yang, E.V. & Glaser, R. (2002) Stress-associated immunomodulation and its implications for responses tovaccination. Expert Review of Vaccines 1,453–459.

In: Psychoneuroimmunology, 3rd edn.(Ader, R., Felten, D.L. & Cohen, N.,eds.). Academic Press, San Diego, CA:565–582.

Tvede, N., Heilmann, C., Halkjaer-Kristensen, J. & Pedersen, B.K. (1989).Mechanisms of B-lymphocytesuppression induced by acute physicalexercise. Journal of Clinical LaboratoryMedicine 30, 169–173.

Verloop, J., Rookus, M.A., van der Koou,K. & van Leeuven, F.E. (2000) Physicalactivity and breast cancer risk in womenaged 20–54 years. Journal of the NationalCancer Institute 92, 128–135.

Vilcek, J. (2003) The cytokines: anoverview. In: The Cytokine Handbook, 4thedn. (Thomson, A.W. & Lotze, M.T.,eds.). Academic Press, New York: 3–17.

Weidner, T. & Schurr, T. (2003) Effect ofexercise on upper respiratory tractinfection in sedentary subjects. BritishJournal of Sports Medicine 37(4), 304–306.

Weidner, T.G., Cranston, T., Swchurr, T. & Kaminsky, L.A. (1998) The effect ofexercise training on the severity and

Page 404: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

388

Introduction

Insulin stimulates pleiotropic pathways resulting in increases in both oxidative and non-oxidative glucose disposal, protein synthesis, gene transcrip-tion and cellular growth and hypertrophy. It haslong been known that exercise is able to elicit similareffects in skeletal muscle. A single bout of exercisecan have profound effects on substrate metabolism,and chronic exercise is associated with adaptationsthat enhance mechanical and metabolic efficiency in skeletal muscle. Among these adaptations arechanges in glucose and glycogen metabolism, pro-tein synthesis and hypertrophy, as well as changesin gene transcription.

Efforts have been made to uncover the mechan-isms by which exercise is able to mimic and enhancespecific effects of insulin, many of which result inclinically relevant adaptations. A fundamental issuein understanding the biological effects of exercise inskeletal muscle is elucidation of intracellular signal-ing mechanisms that enable exercise to enhanceinsulin responsiveness. Interestingly, while it hasbeen recognized that insulin and exercise utilize dis-tinct signaling pathways leading to various cellulareffects, more recent evidence has emerged showingthat exercise and insulin utilize similar signalingintermediates as well.

This chapter will highlight how insulin regulatesglucose transport, glycogen metabolism and proteinmetabolism utilizing the classical insulin-stimulatedphosphatidylinositol 3-kinase (PI3K) pathway. Wewill then review how exercise is able to mimic manyof these metabolic effects in skeletal muscle utilizing

alternative signaling cascades like the PI3K-inde-pendent 5′-AMP-activated protein kinase (AMPK)pathway. While insulin and exercise have beenshown to regulate independent pathways leading tothe regulation of certain metabolic processes (i.e.glucose transport), we will also discuss how exer-cise and insulin are both able to regulate commonsignaling pathways like the family of mitogen-activated protein kinases (MAPKs). Finally, we willdiscuss the clinical relevance of the various effects ofexercise on insulin action in skeletal muscle.

Insulin action in skeletal muscle

Glucose transport

Insulin stimulation has long been known to result inincreases in GLUT4 translocation, glucose trans-port, glycogen synthesis and protein synthesis inskeletal muscle. Insulin elicits many of its metaboliceffects in skeletal muscle by activation of the class-ical PI3K pathway (Fig. 27.1). Under normal phy-siologic conditions, insulin binds to the α subunit of the insulin receptor (IR). This binding activatesautophosphorylation activity in the β subunit andsubsequently causes tyrosine phosphorylation ofnumerous insulin receptor substrates. For example,once activated, the pleckstrin homology domain ofthe insulin receptor substrate-1 and -2 (IRS-1/-2)can dock with the p85 regulatory subunit of PI3Kand activate its p110 catalytic subunit. Catalyticactivity of PI3K results in the phosphorylation ofphosphoinositide (PI) 4,5-bisphosphate to PI 3,4,5-trisphosphate (PIP3). PIP3 is necessary to activate

Chapter 27

Exercise Regulation of Insulin Action inSkeletal Muscle

RICHARD C. HO, OSCAR ALCAZAR AND LAURIE J. GOODYEAR

Page 405: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 389

3-phosphoinositide-dependent protein kinase-1(PDK1), which phosphorylates Akt (also known asPKB) on threonine 308. Subsequent phosphoryla-tion of Akt on serine 473 by a yet uncharacterizedkinase further activates the enzyme. Glycogen syn-thase kinase-3 (GSK3), the mammalian target ofrapamycin (mTOR), and the 70-kDa S6 proteinkinase (S6K) are among the established downstreamsubstrates of Akt. Signaling through this classicalPI3K pathway leads to increases in glucose uptakevia the translocation of the insulin-sensitive GLUT4glucose transporter from subcellular vesicles to theplasma membrane. AS160 has recently been pro-posed to be an Akt-targeted signaling protein medi-ating GLUT4 translocation (Sano et al. 2003).

Glycogen synthesis

The ability of insulin to stimulate glycogen syn-thesis was believed to involve the activation of pro-tein phosphatase-1 (PP1), and deactivation of GSK3(Cross et al. 1995, 1997; Brady et al. 1998; Liu &Brautigan 2000). Through its interaction with glyco-gen-targeting subunits, insulin activates PP1, whichcatalyzes the dephosphorlyation and activation of glycogen phosphorylase. However, mice lackingthe regulatory subunit of muscle-specific PP1 (PP1G)exhibit normal insulin-stimulated glycogen synthaseactivation (Suzuki et al. 2001), suggesting the involve-ment of alternative pathways. Under basal condi-tions, active GSK3 exists in a non-phosphorylated

Glucosetransport

Glycogensynthesis

Proteinsynthesis

PDK1

PIP3

p110

p85 IRS

Akt mTOR

4E-BP1 S6K

elF4E

GSK3

PP1 Glycogensynthase

AS160

?

GLUT4translocation

GLUT4

Insulinreceptor

α subunit

β subunit

Fig. 27.1 Insulin signaling throughthe classical phosphatidylinositol 3-kinase (PI3K) pathway. Insulinbinding to the extracellular insulinreceptor α subunit activates catalyticactivity in the correspondingtransmembrane β subunit. The β subunit catalyzes thephosphorylation of numerous insulinreceptor substrates, some of which(e.g. IRS-1/-2) associate with andactivate the p85 and p110 subunits ofPI3K. Activated PI3K results in theproduction of 3,4,5-trisphosphate(PIP3), and serial activation of 3-phosphoinositide-dependent proteinkinase-1 (PDK1) and Akt. Aktsignaling bifurcates resulting in themodification of proteins involved inthe regulation of glucose transportertranslocation, glycogen synthesis andprotein synthesis.

Page 406: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

390 chapter 27

state. Active GSK3 phosphorylates and inhibitsglycogen synthase activity. Upon stimulation byinsulin, Akt catalyzes the phosphorylation of GSK3,converting it from its active to inactive form. This, inturn, relieves the negative regulation that GSK3exerts on glycogen synthase and thus promotesglycogen accretion (Cross et al. 1995). Evidence forinsulin-induced deactivation of GSK3 has beenshown in both muscle cells (Cross 1995) and skeletalmuscle tissue (Markuns et al. 1999; Wojtaszewski et al. 2000).

Protein synthesis

The function of insulin and its mitogenic andanabolic properties in various tissues has been wellcharacterized. Several lines of evidence have sug-gested that post-prandial increases in insulin areassociated with increased protein synthetic rates inskeletal muscle (Shah et al. 2000). Studies have sug-gested that insulin-stimulated increases in rates ofprotein synthesis are mediated by PI3K-dependentmechanisms, since PI3K inhibitors can attenuate theactivation of key regulatory molecules involved inprotein synthesis. Presumably, this mechanism in-volves the sequential activation of PI3K, Akt, mTORand S6K, and deactivation of eukaryotic initiationfactor 4E binding protein 1 (4E-BP1) (reviewed inShah et al. 2000; Kimball et al. 2002). The ability of insulin to promote protein synthesis in skel-etal muscle is also partially mediated by PI3K-dependent signaling to eukaryotic initiation factor2B (eIF2B), a guanine nucleotide exchange proteininvolved in the regulation of mRNA translation ini-tiation (Welsh et al. 1998). Existing data suggest thatunder non-stimulated conditions, GSK3 phosphory-lates eIF2B on an inhibitory serine residue (Welsh et al. 1998). Acute insulin stimulation leads to thephosphorylation and inactivation of GSK3, result-ing in the activation of eIF2B in skeletal muscle.

Exercise mimics insulin action inskeletal muscle

Like insulin, exercise is able to stimulate GLUT4translocation, glucose transport, glycogen synthesisand protein synthesis in skeletal muscle. Therefore,

it was a logical hypothesis that exercise and insulinutilized similar signaling cascades in skeletal muscle.However, several studies have clearly demonstratedthat proximal insulin-stimulated PI3K intermedi-ates are not involved in the mechanism by whichexercise elicits its metabolic effects. For example,contractile activity does not lead to increases inautophosphorylation of isolated insulin receptors(Treadway et al. 1989) or IRS-1 and IRS-2 tyrosinephosphorylation (Goodyear et al. 1995; Sherwood et al. 1999; Wojtaszewski et al. 1999a; Howlett et al.2002). Consistent with this, muscle-specific insulinreceptor knockout mice exhibit normal exercise-mediated glucose transport (Wojtaszewski et al.1999a). PI3K activity has been reported to beunchanged with skeletal muscle contraction (Good-year et al. 1995; Wojtaszewski et al. 1996, 1999a).Additionally, wortmannin, a PI3K inhibitor, doesnot impair contraction-stimulated glucose transport(Hayashi et al. 1998). The lack of activation of theseproximal PI3K signaling intermediates demon-strates that the underlying molecular mechanismsleading to the insulin- and contraction-inducedstimulation of glucose uptake and glycogen syn-thesis in skeletal muscle are distinct. This is alsosupported by evidence showing that the effects ofacute exercise and insulin on glucose transport areadditive (Ruderman et al. 1971).

Controversy exists regarding the potential role ofAkt in contraction-mediated signaling in skeletalmuscle. While early studies reported that exerciseand contraction did not result in the activation of Akt (Brozinick & Birnhaum 1998; Lund et al. 1998; Widegren et al. 1998; Sherwood et al. 1999;Wojtaszewski et al. 1999a), other studies haveshown significant activation or phosphorylation of Akt in intact skeletal muscles in response to con-traction (Turinsky & Damrau-Abney 1999; Nader &Esser 2001). Recently, we and others have foundthat both exercise in vivo and contraction in situ viasciatic nerve stimulation increases Akt phosphory-lation in multiple rat hindlimb muscles (Turinsky & Damrau-Abrey 1999; Sakamoto et al. 2002, 2003).It has also been demonstrated that, like insulin,exercise alters the activity of GSK3 in rat skeletalmuscles (Markuns et al. 1999). Insulin and exerciseincreased glycogen synthase activity to a similar

Page 407: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 391

extent, however, the mechanisms involved have beenshown to be slightly different. Exercise deactivatesGSK3α and β activity, and increases phosphoryla-tion to a similar degree as does insulin (Sakamoto et al. 2003). Another study showed that bicycle exercise increased glycogen synthase activity imme-diately following exercise without detectable deac-tivation of either isoform of GSK3 in vastus lateralismuscle (Wojtaszewski et al. 2001). There may bealternative mechanisms in the regulation of GSK3activity in skeletal muscle. Interestingly, we haverecently found that the muscle-specific regulatorysubunit of PP1 is required for regulation of glycogenmetabolism under basal conditions and in responseto contractile, but not insulin, activity (Aschenbachet al. 2001). Therefore, insulin-stimulated glycogensynthase activation seems to be regulated by GSK3,while exercise may regulate glycogen synthase viaGSK3-independent mechanisms.

Exercise-stimulated AMPK activation

Since PI3K appears not to be required for exercise-mediated glucose transport in skeletal muscle, anindependent pathway activated by exercise hasbeen the focus of much attention. The discovery of the AMPK as an enzyme potently regulated byexercise has been revealed as such a pathway. As a member of a metabolite-sensing protein kinasefamily, AMPK acts as a fuel gauge monitoring cellu-lar energy levels. Under conditions of decreased cel-lular energy status (increase in the AMP/ATP and creatine/phosphocreatine ratios), AMPK signalingfunctions to down-regulate adenosine triphosphate(ATP)-consuming pathways and up-regulate altern-ative pathways for ATP regeneration (Fig. 27.2).Skeletal muscle contraction is known to depletethese intracellular energy stores, and consequently,AMPK has been shown to exhibit robust activa-tion in response to both exercise and contraction(Winder & Hardie 1996; Rasmussen & Winder 1997;Vavvas et al. 1997). AMPK is activated by a complexmechanism that involves allosteric modification,decreases in phosphatase activities and phosphory-lation by an AMPK recently identified to be LKB1(Hawley et al. 2003; Ossipova et al. 2003; Lizcano et al. 2004).

Early evidence suggested a role for AMPK in theregulation of fat oxidation (Vavvas et al. 1997;Hardie et al. 1998). Recently, studies from our labor-atory (Hayashi et al. 1998, 2000) and others (Hardieet al. 1998; Russell et al. 1999) have also described arole for AMPK in mediating contraction-inducedglucose transport. Both contraction and the AMPKactivator 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) stimulate glucose trans-port by insulin-independent mechanisms (Hayashiet al. 1998), and AMPK activity is associated withglucose transport in contracting skeletal muscle(Ihlemann et al. 1999). Mice overexpressing a dominant inhibitory AMPK mutant were shown toexhibit complete inhibition of AICAR-mediatedglucose uptake, but only partial inhibition of contraction-mediated glucose uptake into skeletalmuscle (Mu et al. 2001). There are two isoforms ofthe AMPK catalytic subunit (α1 and α2), and anotherstudy has recently demonstrated that AMPK α2knockout mice exhibit significant impairments inAICAR-mediated, yet completely normal skeletalmuscle glucose transport, in response to contrac-tion (Jorgensen et al. 2004). These data indicate thatwhile AMPK is required for glucose transport inresponse to AICAR, alternative exercise-responsivesignaling pathways are involved in contractingskeletal muscle.

Recent studies suggest that AMPK is involved inthe regulation of glycogen metabolism, althoughexisting data are equivocal, suggesting both aninhibitory and activating role of AMPK in the regu-lation of glycogen synthesis. For example, somestudies proposed that AMPK has the ability to phos-phorylate key proteins of glycogen metabolism invitro, such as glycogen synthase (Carling & Hardie1989), which would be expected to inhibit glycogensynthesis (Skurat et al. 1994) and phosphorylasekinase (Carling & Hardie 1989), the upstream effec-tor of glycogen phosphorylase. It seems reasonableto speculate that a major role of AMPK in contract-ing muscle would be to promote glycogen degrada-tion and inhibit glycogen synthesis, as AMPK isknown to buffer intracellular ATP levels in variouscell systems. In accordance with this hypothesis,there is a study showing an increased glycogen con-tent in skeletal muscle of Hampshire pigs, which

Page 408: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

392 chapter 27

harbor a point mutation in the γ3 subunit of AMPKthat renders this enzyme less active (Milan et al. 2000).In contrast to this, reports have emerged showingthat similar mutations in the γ1 and γ2 subunits resultin constitutively active AMPK in conjunction withelevated muscle glycogen levels (Hamilton et al.2001; Arad et al. 2002). Pharmacological activation of AMPK by AICAR in hindlimb muscles has beenshown to result in the phosphorylation and deac-tivation of glycogen synthase in rat soleus, and redand white gastrocnemius muscles (Wojtaszewski et al. 2002). In contrast, it has also been reported thatAMPK functions to increase glycogen synthesiswith chronic AICAR treatment in red, slow-twitchand white, fast-twitch quadriceps and gastrocne-mius muscles (Winder et al. 2000; Buhl et al. 2001).

Exercise is able to regulate glucose transport and glycogen synthesis in skeletal muscle utilizing

insulin-independent pathways (Wallberg-Henriksson& Holloszy 1984, 1985). Interestingly, while oper-ating through distinct signaling cascades during exercise, the post-exercise period is characterized by enhancements in insulin sensitivity and respons-iveness. These effects of exercise on insulin actionand skeletal muscle metabolism persist well into thepost-exercise period.

Insulin sensitivity: acute exercise increasesinsulin action on glucose disposal

In addition to the well-characterized increase in glucose transport during exercise, prior exercise has also been reported to improve insulin action in skeletal muscle. Richter et al. (1982) made the ini-tial observation that previously exercised musclesexhibit elevated insulin-stimulated glucose uptake,

EXERCISE

Glucosetransport

Fatoxidation

Mitochondrialbiogenesis

Proteinsynthesis

Genetranscription

LKB1

AMPK

PGC-1

ACC

CPT-1

?

?

?

?

?GLUT4translocation

Malonyl CoA

GLUT4

Fig. 27.2 Exercise regulation of 5′-AMP-activated protein kinase(AMPK) in skeletal muscle. Exerciseactivates AMPK through changes in cellular energy status (i.e. AMP : ATP), and potentially throughthe recently described AMPK, LKB1.AMPK is known to inhibit ATP-consuming pathways such as proteinsynthesis, and activate adenosinetriphosphate (ATP) regeneratingpathways such as fat oxidation.AMPK is also believed to regulatemitochondrial biogenesis via PGC-1α, as well as additional cellularprocesses such as glucose transportand gene transcription and throughmechanisms that have not been fully elucidated. ACC, acetyl-CoAcarboxylase.

Page 409: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 393

even when the effects of the exercise session per se are no longer present. This finding has beenverified in numerous animal and human studiesusing a variety of experimental models. In humans,an increase in insulin-stimulated whole body gluc-ose utilization after exercise has been observed(Bogardus et al. 1983; Devlin et al. 1987; Mikines et al.1988; Richter et al. 1989), and by using the arterioven-ous balance technique this effect was shown to be primarily mediated by an increase in skeletalmuscle glucose disposal (Ivy & Holloszy 1981;Richter et al. 1982).

An early study using a rat model first suggestedthat the increase in insulin sensitivity with exer-cise is restricted to the working muscle (Richter et al. 1984). One-legged exercise models in humanshave also demonstrated that the exercise-inducedincrease in insulin-stimulated glucose uptake is a local phenomenon restricted to the exercised mus-cles, and strongly suggested that changes in sys-temic factors are not the cause of the increase ininsulin sensitivity to stimulate muscle glucoseuptake (Richter et al. 1984, 1989). Interestingly, if isolated epithroclearis muscles from rats are con-tracted by applying electrical stimulation in vitro, no changes in insulin sensitivity are observed following the contraction period (Wardzala et al.1985). This observation and a subsequent study(Kolterman et al. 1980) led some to suggest that a fac-tor released into the circulation during contractileactivity is necessary for the post-exercise increase in insulin sensitivity. Since there is an increase ininsulin sensitivity to stimulate glucose transport inmuscles of the perfused rat hindlimb after electricalstimulation (Richter et al. 1984), it might be the para-crine action of a neurotrophic factor that initiates theevents leading to the increase in insulin sensitivityafter exercise (Sasson et al. 1987).

Exercise results in increases in blood flow toworking muscles, and it has been hypothesized thatdifferences in insulin delivery between exercisedand rested muscles could explain the enhancedinsulin action observed in the post-exercise period.However, using a euglycemic–hyperinsulinemicclamp, studies have shown that exercised legsexhibit higher insulin sensitivity compared withnon-exercised legs, even when no differences are

observed between insulin delivery and clearance(Bogardus et al. 1983; Devlin & Horton 1985; Richteret al. 1989). Furthermore, studies using isolatedmuscles (in vitro or perfused) where insulin levelsare controlled show that insulin sensitivity isincreased with exercise (Ruderman et al. 1971).Additionally, the majority of studies show that exercise does not increase the binding of insulin toits receptor (Bonen et al. 1984; Zorzano et al. 1985;Treadway et al. 1989). Therefore, neither higherinsulin delivery nor insulin binding to the receptorexplains the increase in insulin action in previouslyexercised skeletal muscles. Taken together, thesedata suggest that modulation of a post-insulinreceptor event may be involved in the increasedinsulin action after exercise.

The cellular mechanisms leading to the increasein insulin sensitivity following exercise have beenhypothesized to involve enhanced insulin signal-ing. However, as discussed earlier, studies haveshown that exercise does not utilize proximal PI3Ksignaling molecules during exercise, yet specula-tions were made that exercise resulted in enhance-ments in insulin-stimulated PI3K signaling in thepost-exercise period. Prior exercise does not changeinsulin binding to its receptor (Bonen et al. 1984;Zorzano et al. 1985; Treadway et al. 1989) or increaseinsulin-stimulated receptor tyrosine kinase activityin skeletal muscles obtained from rats (Treadway et al. 1989; Goodyear et al. 1995) or humans(Wojtaszewski et al. 2000). In fact, prior contractionof rat hindlimb skeletal muscles has been shown tocause a paradoxical decrease in insulin-stimulatedtyrosine phosphorylation of IRS1 and IRS1-associatedPI3K activity (Wojtaszewski et al. 2000).

In contrast to these reports, other evidence sug-gests that the increase in insulin responsiveness to stimulate muscle glucose transport immediatelyfollowing exercise is associated with an increase in insulin-stimulated PI3K activity (Houmard et al.1999; Chibalin et al. 2000; Kirwan et al. 2000).Furthermore, we have reported that prior exerciseresulted in enhanced insulin-stimulated IRS-2-associated PI3K activity compared with insulinstimulation alone (Howlett et al. 2002). The increasein insulin-stimulated PI3K activity following exer-cise is short-lived, since it is not present when

Page 410: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

394 chapter 27

insulin stimulation occurs after 30 min of recoveryfrom exercise (J.F. Wojtaszewski & L.J. Goodyear,unpublished observation). Furthermore, if a physio-logical hyperinsulinemic clamp is applied 3 h afterone-legged exercise in humans, PI3K activity is not higher in the muscle from the exercised leg(Wojtaszewski et al. 1997). The lack of enhancementin insulin-stimulated PI3K activity in these latertime points after exercise is consistent with findingsin humans showing no change in insulin receptortyrosine kinase activity or IRS-1 tyrosine phospho-rylation showing no change in insulin receptor tyrosine kinase activity or IRS-1 tyrosine phospho-rylation (Wojtaszewski et al. 1997). Thus, althoughthere seems to be an up-regulation of some com-ponents of the insulin signaling pathway withinsulin immediately after exercise, these changes donot explain the long-lasting influence of exercise oninsulin action in skeletal muscle. These studies ruleout a role for enhanced insulin signaling as a mech-anism for increased glucose uptake after an acuteexercise bout, and provide additional support to the hypothesis that exercise and insulin act throughdistinct signaling mechanisms.

Elevations in post-exercise insulin sensitivity arealso associated with the degree of muscle glycogendepletion. It is generally known that increases ininsulin sensitivity following exercise can persist ifmuscle glycogen levels are kept low. Muscle gluc-ose uptake following exercise remains elevated forup to 18 h if rats are fed a carbohydrate-free diet,while rates of uptake return to pre-exercise levels ifcarbohydrate is provided following exercise (Younget al. 1983).

Exercise training increases insulin action onglucose disposal

While the effects of an acute bout of exercise oninsulin-stimulated glucose transport in the post-exercise period have been well established, theseeffects are quite transient with improvements ininsulin responsiveness returning to pre-exerciselevels, usually within 12 h of the preceding exercisebout. It was reasonable to speculate that chronicexercise would result in adaptations that enableskeletal muscle to exhibit more sustained increases

in insulin action. Three decades ago, Bjorntorp et al. (1972) suggested that exercise training mightincrease tissue sensitivity to insulin. This investiga-tion, along with several subsequent studies (Lohmanet al. 1978; Johansen & Munch 1979; LeBlanc et al.1979, 1981; Seals et al. 1984), demonstrated that compared with sedentary individuals, trained indi-viduals tend to exhibit improvements in glucose tolerance and insulin sensitivity. Additional studiesusing the hyperinsulinemic–euglycemic clamp havedemonstrated that exercise-trained people havehigher rates of insulin-stimulated glucose disposalthan do their sedentary counterparts (Saltin et al.1978; Rosenthal et al. 1983; Hollenbeck et al. 1984;King et al. 1987; Mikines et al. 1989). Although theseresults have been interpreted to indicate that exer-cise training results in an increase in tissue sensitiv-ity to insulin, this concept has been complicated by the fact that, as discussed previously, an acutebout of exercise produces major effects on bothwhole body glucose disposal and metabolism(Pruett & Oseid 1970; Bogardus et al. 1983; Devlin etal. 1985, 1987; Mikines et al. 1988, 1989) and skeletalmuscle glucose uptake and metabolism (Holloszy & Narahara 1965; Ivy & Hollozy 1981; Elbrink &Phipps 1980; Fell et al. 1982; Richter et al. 1989) thatcan persist well into the post-exercise period. Hencemany of the effects of regular exercise may be due tothe residual effects of the last individual exercisesession, rather than long-term adaptations to exer-cise training. Several studies have attempted to dis-criminate differences in insulin sensitivity betweenacute exercise effects and training adaptations(Heath et al. 1983; Burstein et al. 1985; King et al.1988; Mikines et al. 1989). Some of these studies suggest that the increases in insulin sensitivity asso-ciated with exercise training are transient (Bursteinet al. 1985; King et al. 1988). One study has reportedthat while 10 days of detraining resulted indecreases in insulin sensitivity to a level comparablewith sedentary individuals, performance of a singleexercise bout by the trained subjects on the 11th dayof the protocol did not completely return the insulinresponse to the trained level (Heath et al. 1983).These data suggest that effects of an acute bout of exercise cannot completely account for increasesin insulin action associated with exercise training.

Page 411: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 395

After comparing trained subjects with untrainedsubjects who had undergone an acute bout of exer-cise and after comparing trained subjects before and after 5 days of detraining (Mikines et al. 1989), adifferent group of investigators concluded that anincrease in maximal insulin action on whole-bodyglucose uptake (insulin responsiveness) but notinsulin sensitivity is a long-term adaptation causedby endurance exercise training. Additional studieshave reported that maximal insulin-stimulated gluc-ose disposal was unaffected by 10 days of inactivity(King et al. 1988), suggesting that the reduction ininsulin action following short-term inactivity is theresult of a decrease in insulin sensitivity and not adecrease in insulin responsiveness. The molecularbasis for this phenomenon has not been completelyelucidated, but appears to be dependent on multiplefactors including humoral factors, muscle glyco-gen concentrations and alterations in signalingmechanisms.

One study has recently reported that habitualexercise was associated with decreased proteinexpression of IR, IRS-1 and IRS-2 in trained versusuntrained subjects (Yu et al. 2001). Another studyfound that 7 days of exercise training did not elicitchanges in PI3K activity, despite increases in insulinaction in skeletal muscle (Tanner et al. 2002). Theenhanced activation by insulin was not associatedwith a greater insulin-stimulated insulin receptor or IRS-1 tyrosine phosphorylation. These data suggest that the improvement in insulin action associated with exercise training is not associatedwith increases in PI3K signaling. Contrary to thesedata, trained rats have been shown to exhibit higherinsulin-stimulated glucose transport, IRS-1/-2 activ-ity, PI3K association and Akt phosphorylation com-pared with sedentary controls (Chibalin et al. 2000;Luciano et al. 2002). Therefore, increases in insulinresponsiveness resulting from chronic exercise may be mediated, in part by alterations in proximalPI3K signaling in skeletal muscle, but most likelyinvolves additional signaling molecules whosecharacterization remains elusive.

Efficacious exercise training programs can resultin changes in plasma lipids and body composi-tion, in particular, decreases in total body fat andincreases in lean body mass. These changes, inde-

pendent of short-term exercise effects are associatedwith improvements of insulin action in skeletalmuscle. However, exercise-mediated improve-ments in insulin sensitivity have been detected evenwhen studies have controlled for these potentiallyconfounding effects (Oshida et al. 1989; Hughes et al.1993). Hence, exercise training appears to improveinsulin sensitivity independent of changes in bodycomposition and there may be additive effects ofexercise and decreased adiposity.

Glycogen supercompensation: exercise increasesinsulin action on glycogen metabolism

The increase in glycogen synthesis in response toinsulin stimulation has been well characterized.Furthermore, exercise elicits a net breakdown ofmuscle glycogen despite increases in glycogen syn-thase activity. In 1966, Bergstrom and Hultman(1966) first described the concept of glycogen super-compensation, whereby glycogen levels increasesignificantly in response to carbohydrate feedingfollowing a glycogen depleting exercise bout. Thepreceding exercise bout leads to increases in skeletalmuscle glycogen above that which is seen followinga meal. This effect is mediated to a large extent bythe post-exercise increases in glucose transport,glycogen synthase activity and the degree of glyco-gen depletion.

Increases in post-exercise glucose transport resultin subsequent elevations in the levels of glucose 6-phosphate (G6P), an allosteric activator of glycogensynthase (Price et al. 2003). Therefore, the extent towhich an exercise bout increases glucose transportin the post-exercise period partially determines levels of glycogen synthesis. However, studies haveshown that increases in glycogen synthase activityfollowing exercise cannot fully account for theobserved elevations in glycogen accumulation(Bergstrom et al. 1972; Conlee et al. 1978; Nakatani et al. 1997; Greiwe et al. 1999).

The degree of glycogen depletion resulting fromthe preceding exercise bout is a critical factor indetermining the subsequent increases in muscleglucose uptake and glycogen synthesis followingexercise. In fact, the potentiating effect that exerciseexerts on insulin action (glucose transport, glycogen

Page 412: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

396 chapter 27

synthesis) is a function of the degree of glycogendepletion achieved during the bout of exercise.Interestingly, post-exercise increases in glucoseuptake and insulin sensitivity can be reversed withcarbohydrate feeding and glycogen repletion (Hostet al. 1998; Kawanaka et al. 2000). Similarly, main-taining low glycogen levels following exercise bycarbohydrate restriction extends the effect of exer-cise on insulin action in skeletal muscle (Garcia-Roves et al. 2003). However, a study performed inindividual tissues from perfused hindquarter mus-cle suggests that glycogen is not the only regulatorin the supercompensation phenomenon (Zorzano et al. 1986). In this report, glucose transport returnedto near baseline values even though glycogen con-centrations were maintained at a low level. Thisresult suggests the existence of alternate com-pensatory mechanisms that may participate in theregulation of the supercompensation phenomenontogether with the glycogen levels. The precise mechanism(s) involved in the supercompensationeffect remains to be elucidated. A recent study usingAICAR treatment as a pharmacological model tomimic exercise, in fast-twitch (epitrochlearis) andslow-twitch (flexor digitorum brevis) muscles, re-vealed that AICAR has no effect on either glycogensynthase or glycogen phosphorylase in both muscletypes (Aschenbach et al. 2002). Therefore, this studysuggests that AMPK does not directly regulateglycogen synthase or glycogen phosphorylase inskeletal muscle. Consistent with this finding, a recentinvestigation using four different phosphorylasekinase substrates demonstrated that glycogen phos-phorylase is not a substrate of AMPK in vitro (Beyeret al. 2000). Further efforts are required in the futureto investigate the supercompensation mechanismand its regulation.

Hypertrophy: exercise increases insulin action onprotein synthesis

While skeletal muscle is capable of respondingacutely to changes in circulating humoral factors(insulin) and metabolic demands (exercise), skeletalmuscle also exhibits the ability to adapt to chronicperturbations in such a way that size (hypertrophy),mechanical function (force generation) and meta-

bolic function (oxidative capacity) improve. In particular, resistance exercise is associated with anup-regulation of anabolic processes that allow forskeletal muscle growth and regeneration. One of themain underlying factors responsible for these adap-tations is the ability of exercise to elicit transientincreases in protein synthesis (Booth & Thomason1991; Fluckey et al. 1996). A protocol involving 4days of resistance exercise resulted in elevations ininsulin-mediated protein synthesis in rat skeletalmuscle (Fluckey et al. 1996). Other studies havereported no such effect following endurance exer-cise (Dohm et al. 1980; Balon et al. 1990). This maypartially explain why chronic resistance exerciseresults in muscle hypertrophy, while chronicendurance exercise does not. AMPK functions todown-regulate energy consuming pathways, whileup-regulating ATP-regenerating pathways. Proteinsynthesis is an expensive energy consuming pro-cess, and subsequently, studies have shown thatAMPK is involved in the inhibition of protein syn-thetic pathways (Bolster et al. 2002; Horman et al.2002; Krause et al. 2002; Kimura et al. 2003). There-fore, increases in protein synthesis in skeletal musclefollowing exercise appear to occur via AMPK-independent mechanisms.

The capacity of exercise to mimic the anaboliceffects of insulin has been attributed to the activa-tion of similar signaling mechanisms. For example,studies have demonstrated that exercise activatesMAPK, Akt, S6K and eIF2B, all of which have been implicated in the regulation of skeletal muscleprotein synthesis by insulin (Baar & Esser 1999;Farrell et al. 2000; Sakamoto & Goodyear 2002).Interestingly, exercise and insulin exhibit a synergy,since studies have reported that the ability of resist-ance exercise to increase protein synthesis (andeIF2B activity) in skeletal muscle is dependent on permissive levels of insulin (Fedele et al. 2000;Kostyak et al. 2001). Under moderately hypo-insulinemic conditions, resistance exercise has beenreported to stimulate protein synthesis through a mechanism believed to involve compensatoryincreases in intramuscular IGF-1 signaling (Farrellet al. 1999; Fedele et al. 2000).

While resistance exercise is known to activateenzymes involved in protein synthesis, maximal

Page 413: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 397

protein accretion is highly dependent on additionalfactors. Exercise is able to improve the anaboliceffects of insulin in skeletal muscle, and interactionswith other hormones (e.g. insulin-like growth factorI [IGF-I], testosterone) and substrate availability(amino acids in particular) determine to a largeextent subsequent degrees of hypertrophy (reviewedin Tipton & Wolfe 2001).

Caveat: exercise-induced muscle injury candecrease insulin action on glucose disposal

As described above, exercise is generally associatedwith improvements in insulin-mediated glucosedisposal in skeletal muscle. However, acute decreasesin insulin-stimulated glucose metabolism have been documented following resistance exercise, aphenomenon that is largely explained by the degreeto which the muscle is damaged. The associationbetween whole body injury, trauma and skeletalmuscle insulin resistance has been well documented(Black et al. 1982). Exercise resulting in muscle damage elicits an inflammatory response, wherebyaccumulation of inflammatory cells invades theaffected tissues (Asp et al. 1997). These cells facilitatein the repair, remodeling and removal of damagedtissue. The release of inflammatory cytokines medi-ate many of these repair mechanisms, however, theability of cytokines (i.e. tumor necrosis factor-α) toinduce insulin resistance is also well known.

Due to force/area dynamics, muscle injury ismore likely to occur in response to eccentric (length-ening of contracting muscle) compared with con-centric (shortening of contracting muscle) musclecontraction. While a single bout of concentric exer-cise is associated with improvements in skeletalmuscle insulin action (Richter et al. 1982, 1989;Bogardus et al. 1983), a bout of eccentric exercise wasassociated with impaired whole body insulin action48 h post-exercise (Kirwan et al. 1992). This effectwas also observed in isolated muscle, suggestingthat the insulin resistance is a local phenomenon(Asp & Richter 1996). Consistent with differences ininsulin-mediated glucose metabolism, concentricexercise is commonly associated with increases inGLUT4 protein levels (Richter et al. 1989), whileeccentric exercise has been associated with reduc-

tions in GLUT4 transcription, mRNA and proteinexpression in skeletal muscle by up to 65% (Asp et al. 1995; Kristiansen et al. 1996). Interestingly,while studies have shown that eccentric contractioncan impair glucose metabolism, protein syntheticrates remain elevated. This is perhaps due to select-ive impairments in PI3K signaling and GLUT4translocation (Fluckey et al. 1999) but normallyactivated MAPK signaling (Haddad & Addams2002). In fact, MAPKs are not only activated by muscle contraction, but have also been shown to beactivated by stretch, muscle damage and injury andinflammatory cytokines (discussed later).

While acute moderate-intensity exercise andendurance exercise training are associated withincreases in insulin-mediated glucose disposal,resistance exercise has been shown to elicit oppositeeffects. An early study showed that high intensityresistance exercise was associated with reductionsin glucose disposal (Kirwan et al. 1992). Acute resist-ance exercise has been shown to result in lowerinsulin-stimulated glucose uptake compared withsedentary controls (Fluckey et al. 1999). The de-gree of eccentric contraction involved in resistanceversus aerobic exercise explains, to a large degree,observed differences in insulin-stimulated glucosemetabolism. The anaerobic nature of resistanceexercise results in significant decreases in muscleglycogen. Interestingly, in addition to decreases ininsulin-stimulated glucose transport, eccentric exer-cise has been shown to impair insulin-stimulatedincorporation of glucose into glycogen in variousmuscles (Asp et al. 1997). One study reported thateccentric exercise resulted in a 16% decrease in maximal glycogen synthase activity compared withcontrols (Asp & Richter 1996). These impairmentsare often observed in the more glycolytic fast twitchmuscles, which are highly recruited during resist-ance exercise (Asp & Richter 1996).

While muscle damage resulting from a single boutof resistance exercise can result in decreases inskeletal muscle insulin action, other data suggestthat resistance training is associated with improve-ments in insulin-stimulated glucose disposal (Milleret al. 1994; Yaspelkis et al. 2002). Several factors helpto explain the observed inconsistencies. First, resist-ance exercise includes both concentric and eccentric

Page 414: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

398 chapter 27

components of muscle contraction. Resistance exer-cise programs that include more eccentric contrac-tions would presumably lead to increased muscleinjury and decreased insulin-action and vice versa.Furthermore, while an acute bout of eccentric-resistance exercise is associated with impairmentsin glucose metabolism, chronic resistance exerciseelicits skeletal muscle adaptations that improve in-sulin action in skeletal muscle (e.g. GLUT4 expres-sion). Finally, while untoward effects of an acutebout of eccentric exercise are due to local effects(inflammatory response), corresponding changes inbody composition (decreased fat mass, increasedmuscle mass) associated with chronic resistanceexercise training are associated with improvementsin whole-body insulin action.

Insulin and exercise stimulate MAPK signaling

While it is clear that insulin stimulates glucose disposal utilizing the PI3K pathway, and exerciseappears to utilize PI3K-independent pathways (e.g.AMPK), recent evidence suggests that both exerciseand insulin are able to activate common signalingcascades in skeletal muscle. While insulin has longbeen known to activate the family of MAPK, a grow-ing body of evidence suggests that MAPK signaltransduction pathways play an important role inexercise signaling in skeletal muscle. The MAPKsrepresent an important family of signal transduc-tion proteins, which are expressed in all eukaryoticcells (Fig. 27.3). Four MAPK subgroups have beendescribed: (i) the extracellular-signal regulated kin-ases (ERKs); (ii) c-Jun NH2-terminal kinases (JNKs);(iii) p38 MAPK; and (iv) ERK5/big MAP kinase 1(BMK1). ERKs are predominantly activated bygrowth factors, while JNKs and p38 MAPK are col-lectively known as stress-activated protein kinases,and have been implicated in a large number of cel-lular responses, including cell proliferation, dif-ferentiation, hypertrophy, inflammation, apoptosis,carbohydrate metabolism and gene transcription(Force & Bonventre 1998; Sweeney et al. 1999; Kyriakis& Avruch 2001). In 1996, it was first reported thatexercise activates ERK1/2, JNK and p38 signaling inskeletal muscle (Goodyear et al. 1996). Since that

time there has been intense interest in the regulationof these pathways in skeletal muscle.

ERK signaling

Insulin has long been known to be a potent stimu-lator of ERK in skeletal muscle. Additionally, activa-tion of ERK1/2 signaling has been reported in ratskeletal muscle with treadmill running (Goodyear et al. 1996; Nader & Esser 2001), in vitro contraction(Hayashi et al. 1999; Wojtaszewski et al. 1999b, 2000;Ryder et al. 2000; Wretman et al. 2000, 2001), in situcontraction (Sherwood et al. 1999; Martineau &Gardiner 2001; Nader & Esser 2001), muscle over-load (Carlson et al. 2001) and stretch (Boppart et al.2001); in mouse muscle in response to treadmill run-ning (Dufresne et al. 2001); and in human skeletalmuscle in response to cycle ergometer exercise(Aronson et al. 1997a, 1997b; Widegren et al. 1998;Osman et al. 2000) and marathon running (Yu et al.2001). Upstream of ERK1/2, both MEK1/2 and Raf1(Aronson et al. 1997a, 1997b; Sherwood et al. 1999)activities are increased by exercise and contraction.Molecules downstream of ERK1/2 that have beenshown to be activated by exercise include RSK2(Goodyear et al. 1996; Aronson et al. 1997a, 1997b;Sherwood et al. 1999; Krook et al. 2000; Osman et al.2000; Ryder et al. 2000; Yu et al. 2001) and the mito-gen and stress-activated kinase 1/2 (MSK1/2)(Ryder et al. 2000; Yu et al. 2001). MEK1/2 activa-tion is necessary for ERK1/2 activation since theMEK1/2 inhibitor PD98059 inhibits muscle con-traction induced ERK1/2 phosphorylation (Hayashiet al. 1999; Wojtaszewski et al. 1999b, 2000; Ryder et al. 2000) and its downstream substrates RSK2(Hayashi et al. 1999) and MSK1 (Ryder et al. 2000).

With one-legged cycling exercise in human sub-jects, activation of ERK1/2 (Aronson et al. 1997b;Widegren et al. 1998) and its downstream targets(Aronson et al. 1997b; Krook et al. 2000) is observedin the muscle from the exercised leg, but not in theresting leg. These data suggest that stimulation ofERK1/2 signaling in response to exercise in skeletalmuscle is due to primarily a local, tissue-specificphenomenon, rather than a systemic effect. Themolecules involved in this tissue specific stimula-tion remain elusive.

Page 415: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 399

JNK signaling

Unlike ERK, JNK is only modestly activated byinsulin in skeletal muscle, and some reports showno activation (Aronson et al. 1996). Activation of JNK signaling has been reported in rat skeletalmuscle in response to in vitro contractions (Boppartet al. 2001), in situ contractions (Aronson et al. 1997a;Martineau & Gardiner 2001), treadmill runningexercise (Goodyear et al. 1996), muscle overload(Carlson et al. 2001) and mechanical stretch (Boppartet al. 2001). In human subjects, JNK is activated inresponse to cycle ergometer exercise (Aronson et al.

1998), knee extensions resulting in concentric andeccentric contractions of the quadricep muscles(Boppart et al. 1999) and marathon running (Boppartet al. 2000). In contrast to ERK1/2 signaling, activa-tion of the JNK cascade is sustained during in situmuscle contractions, whereas the activation of the ERK cascade is more rapid and transient. Thissuggests that the upstream proteins that regulatethe JNK signaling cascade are distinct from that ofERK1/2 (Aronson et al. 1997a). Due to the lack ofselective cell permeable inhibitors of JNK signalingat this time, little is known about the physiologicalfunction of JNK in contracting skeletal muscle.

Exercise systemic factors

Signalingmessengers

MAP3K

INSULIN

EXERCISE

Exerciseautocrine and

paracrine factors

Proteinkinases

Gene transcription

Transcriptionfactors

GRB2

SOS

Raf

MEK1/2

ERK1/2

MKK4/7

JNK1/2

MKK3/6

MNK1/2 MSK1/2 RSK

ATF-2 CREB CHOP C-Jun C-Fos Elk-1 MEF2

MAPKAPK2/3

p38

Fig. 27.3 Exercise and insulinregulation of mitogen-activatedprotein kinase (MAPK) signaling inskeletal muscle. Insulin potentlystimulates ERK1/2 activity through a GRB2/SOS pathway. Exercise also regulates MAPK activity viaintracellular signaling molecules, as well as systemic andautocrine/paracrine factors.Downstream substrates of MAPKsinclude various protein kinases, aswell as transcription factors.Mechanisms underlying the ability of exercise to induce changes in geneexpression are relatively unknown;however, modulation via MAPKsignaling represent likely candidates.

Page 416: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

400 chapter 27

p38 signaling

As is the case with JNK, skeletal muscle p38 is weakly activated by insulin, if at all. However,p38 is potently activated in rat skeletal muscle inresponse to treadmill running (Goodyear et al. 1996;Nader & Esser 2001), in vitro contractions (Ryder et al. 2000; Somwar et al. 2000; Wretman et al. 2000,2001; Boppart et al. 2001), in situ contractions (Nader& Esser 2001), muscle overload (Carlson et al. 2001)and mechanical stretch (Boppart et al. 2001). It hasalso been shown that p38 signaling is increased inhumans during cycle ergometer exercise (Widegrenet al. 1998) and marathon running (Boppart et al.2000; Yu et al. 2001). Phosphorylation of MAP-KAPK-2, a downstream substrate for p38, is associ-ated with changes in p38 activity during exercise/contraction and is inhibited by the p38 antagonistSB203580 (Krook et al. 2000; Ryder et al. 2000; Yu et al. 2001).

Does exercise-mediated MAPK activationregulate gene transcription in skeletal muscle?

Chronic exercise can result in changes in skeletalmuscle structure and function. Endurance trainingleads to increases in capillary density, increases in oxidative capacity and mitochondrial density,etc. The exact mechanisms regulating the adaptiveresponse of skeletal muscle to repeated bouts ofexercise are unclear; however, MAPKs are known to activate a number of transcription factors in response to both insulin and exercise. Activation of these transcription factors may contribute to the adaptive effects of exercise in skeletal muscle(Widegren et al. 2001). Using various cell systems,activated MAPKs have been shown to translocate to the nucleus and phosphorylate numerous trans-cription factors such as CREB, Elk, ATF2, c-Jun, c-fos, c-Myc, AP-1, MEF2, NFAT and CHOP (Han et al.1997; Gomez et al. 2000; Kyriakis & Avruch 2001;Hazzalin & Mahadevan 2002). Not only can MAPKsregulate gene transcription by direct interaction withtranscription factors, they can also activate otherdownstream substrates such as RSK2, MAPKAPK-2/3 and MSK1/2 that can translocate to the nucleusand phosphorylate numerous transcription factors.

Downstream consequences of exercise-mediatedMAPK signaling remain obscure; however, recentdata are emerging which implicate p38 in adaptiveresponses to increase lipid metabolism in skeletalmuscle. Peroxisome proliferator activated receptors(PPARs) have received much attention as trans-criptional regulators of lipid metabolism in varioustissues. PPARγ is predominantly expressed in adipo-cytes and has been linked to adipocyte differenti-ation. PPARα expression is high in skeletal muscleand has been shown to regulate the expression ofnumerous genes involved in fat oxidation. Interest-ingly, p38 MAPK has been shown to regulate theactivity of PPARα (Barger et al. 2001). Further-more, p38 has also been shown to regulate PPARγcoactivator-1 (PGC-1) (Knutti et al. 2001; Puigserveret al. 2001). PGC-1 has been shown to regulate mito-chondrial biogenesis in skeletal muscle (Puigserver& Spiegelman 2003). Therefore, the adaptive effectsresulting from exercise training to improve fat util-ization could involve exercise-mediated MAPK activ-ity. Activation of these transcription factors leads to the regulation of gene expression; however, thereis a paucity of data regarding the involvement ofskeletal muscle MAPK activation with exercise.

Effect of MAPK activation on substratemetabolism

While the regulation of gene transcription is anestablished function of MAPK, the involvement ofMAPK in the regulation of cellular substrate meta-bolism is unclear. The ERK1/2 signaling cascadewas previously proposed to be involved in the regulation of both glucose transport and glycogenmetabolism (Merrall et al. 1993). However, in subse-quent studies, a MEK inhibitor that blocks activa-tion of ERK1/2 had no effect on insulin-stimulatedglucose uptake in cultured adipocytes (Haruta et al.1995; Tanti et al. 1996) and skeletal muscles (Hayashiet al. 1999; Wojtaszewski et al. 1999b). Thus, there issubstantial evidence that ERK1/2 signaling is notinvolved in the acute regulation of glucose uptake in response to insulin treatment or contraction inskeletal muscle.

The ERK1/2 signaling cascade has also been proposed to regulate insulin-stimulated activation

Page 417: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 401

of glycogen synthase. This hypothesis was based onthe finding that RSK2 could phosphorylate andinactivate GSK3 and phosphorylate and activate theglycogen-bound form of protein phosphatase-1(PP1-G) in vitro, two reported regulators of insulin-stimulated glycogen synthase activity (Dent et al.1990; Sutherland et al. 1993). However, inhibition ofERK signaling by a MEK inhibitor resulted in noinhibition of insulin-stimulated glycogen synthaseactivity (Lazar et al. 1995). More direct evidence camefrom a study using RSK2 knockout mice whichrevealed that RSK2 is not necessary for insulin-stimulated glycogen synthase activation, and infact, the RSK2 null mice had greater increases inmuscle glycogen synthase activity following insulintreatment (Dufresne et al. 2001). However, thesestudies do not rule out a role for RSK2 in the regula-tion of glycogen metabolism in the basal state, as the knockout mice had lower levels of muscle glycogen.

A growing body of literature is emerging implic-ating MAPK pathways in the regulation of lipidmetabolism. Recently, a study has suggested thatERK signaling is associated with the plasma mem-brane fatty acid transporter FAT/CD36 (Todd &Turcotte 2003). Incubation of isolated muscle withPD98059 significantly attenuated the contraction-induced increase in fatty acid uptake in skeletalmuscle. ERK has also been recently suggested to play a role in the activation of HSL in skeletalmuscle watt (Donsmark et al. 2003; Langfort et al.2003; Watt et al. 2003). These data suggest that ERKactivation is involved in both the uptake of fattyacids and hydrolysis of triglyceride in skeletal muscle. Whether chronic exercise improves insulin-stimulated ERK activity in skeletal muscle remainsto be elucidated.

There have been few studies examining theeffects of JNK activation in the regulation of carbo-hydrate metabolism in skeletal muscle. One studydemonstrated that activation of JNK by anisomycin,a protein synthesis inhibitor, mimics insulin’s actionto stimulate glycogen synthesis in mouse skeletalmuscle in vivo (Moxham et al. 1996). Based on theirfindings, this group concluded that JNK stimulatesglycogen synthase activity through the regulation of RSK3 and GSK3. Since exercise and contraction

robustly activate JNK activity, we hypothesized that JNK could be involved in the regulation of contraction-stimulated glycogen synthase activ-ity. Overexpression of wild type JNK1 in skel-etal muscle in vivo dramatically increased basal andcontraction-stimulated JNK activity. However, thisincrease in JNK activity did not enhance basal andcontraction-stimulated glycogen synthase activ-ity in mouse skeletal muscle, suggesting that JNK is not involved in the regulation of contraction-stimulated glycogen synthase activity (Fujii et al.2001). Whether JNK is involved in glucose transport regulation in muscle is not known and will be animportant area for future investigation.

Recent studies have provided evidence that p38 isinvolved in the regulation of contraction-stimulatedglucose uptake in skeletal muscle. Somwar and colleagues demonstrated that p38 activity and glucose uptake were increased in isolated extensordigitorum longus (EDL) muscles with contraction in vitro (Somwar et al. 2000), and the p38 antagon-ist SB203580 abolished the activation of p38 andreduced contraction-stimulated glucose uptake by40–50%. However, p38 inhibitors are known to havenumerous non-specific effects and it is still unclearwhether the attenuation of glucose uptake was dueto inhibition of contraction-stimulated p38 activityor an indirect effect of the compound on other signaling intermediates (Somwar et al. 2000). Wehave recently showed that p38γ, the isoform highlyabundant in skeletal muscle, is a negative regulatorof GLUT4 expression and contraction-stimulatedglucose uptake (Ho et al. 2004).

The impact of exercise on insulin-stimulatedMAPK activity remains to be uncovered. Interest-ingly, cytokines that are released in response tomuscle damage and implicated in the negative re-gulation of insulin-stimulated glucose metabolism(e.g. tumor necrosis factor-α) are also potent stimu-lators of JNK and p38. Additionally, both JNK andp38 activity have been shown to induce impair-ments in insulin-stimulated glucose transport inskeletal muscle. However, because exercise resultsin significant increases in glucose transport in para-llel with MAPK activation, the exact role of MAPK in contracting skeletal muscle warrants furtherinvestigation.

Page 418: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

402 chapter 27

Clinical implications

Throughout this chapter, we have discussed vari-ous effects of exercise on insulin action in skeletalmuscle. Exercise clearly has the ability to improveglucose transport, glycogen synthesis and proteinmetabolism, as well as stimulate adaptive changesthrough gene transcription. While the majority ofstudies reviewed in this chapter involve healthyindividuals, the impact of exercise on intermediarymetabolism also transcends to individuals exhibit-ing metabolic complications. It has long been recog-nized that physical exercise has important benefitsfor people with obesity and diabetes (Trovati et al.1984; Helmrich et al. 1991). However, the transientnature of post-exercise insulin sensitivity limits the beneficial impact of physical activity. Chronicexercise, on the other hand, results in multiple physical and metabolic adaptations. Exercise train-ing improves glucose tolerance and insulin action ininsulin-resistant humans (Hughes et al. 1993) andtype 2 diabetic patients (Dela et al. 1994). Theimprovements in insulin sensitivity seem to be multifaceted, including alterations in body composi-tion, plasma lipid profiles, intracellular signaling

and protein expression. Furthermore, epidemiolo-gical studies have determined that regular physicalexercise can reduce the risk of developing type 2diabetes (Helmrich et al. 1991; Manson et al. 1991,1992).

Summary

Significant advances have been made in recent years in the elucidation of mechanisms by whichexercise increases insulin action in skeletal muscle.Discoveries have been made showing that bothexercise and insulin stimulate increases in glucosetransport, glycogen metabolism, protein synthesisand long-term adaptations (e.g. hypertrophy).Interestingly, these effects are elicited through bothcommon and distinct signaling pathways. Further-more, additive effects of exercise and insulin in the regulation of intermediary metabolism andadaptive responses have a widespread impact inboth health and disease. While exercise training canresult in adaptations to improve performance,chronic physical activity can also prevent or reversemetabolic defects observed in conditions such astype 2 diabetes.

ReferencesArad, M., Benson, D.W., Perez-Atayde,

A.R. et al. (2002) Constitutively activeAMP kinase mutations cause glycogenstorage disease mimicking hypertrophiccardiomyopathy. Journal of ClinicalInvestigation 109, 357–362.

Aronson, D., Fielding, R.A., Violan, A.,Dufresne, S.D. & Goodyear, L.J. (1996)Exercise activates the MAP kinasesignaling cascade in human skeletalmuscle. Diabetes 45, 103A.

Aronson, D., Dufresne, S.D. & Goodyear,L.J. (1997a) Contractile activitystimulates the c-Jun NH2-terminalkinase pathway in rat skeletal muscle.Journal of Biological Chemistry 272, 25 636–25 640.

Aronson, D., Violan, M.A., Dufresne, S.D.et al. (1997b) Exercise stimulates themitogen-activated protein kinasepathway in human skeletal muscle.Journal of Clinical Investigation 99,1251–1257.

Aronson, D., Boppart, M.D., Dufresne,S.D., Fielding, R.A. & Goodyear, L.J.

(1998) Exercise stimulates c-Jun NH2kinase activity and c-Jun transcriptionalactivity in human skeletal muscle.Biochemical and Biophysical ResearchCommunications 251, 106–110.

Aschenbach, W.G., Suzuki, Y., Breeden, K.et al. (2001) The muscle-specific proteinphosphatase PP1G/RGL(GM) isessential for activation of glycogensynthase by exercise. Journal of BiologicalChemistry 276, 39 959–39 967.

Aschenbach, W.G., Hirshman, M.F., Fujii,N. et al. (2002) Effect of AICAR treatmenton glycogen metabolism in skeletalmuscle. Diabetes 51, 567–573.

Asp, S. & Richter, E.A. (1996) Decreasedinsulin action on muscle glucosetransport after eccentric exercise in rats. Journal of Applied Physiology 81,1924–1928.

Asp, S., Daugaard, J.R. & Richter, E.A.(1995) Eccentric exercise decreasesglucose transporter GLUT4 protein inhuman skeletal muscle. Journal ofPhysiology 482, 705–712.

Asp, S., Watkinson, A., Oakes, N.D. &Kraegen, E.W. (1997) Prior eccentriccontractions impair maximal insulinaction on muscle glucose uptake in the conscious rat. Journal of AppliedPhysiology 82, 1327–1332.

Baar, K. & Esser, K. (1999) Phosphorylationof p70(S6k) correlates with increasedskeletal muscle mass followingresistance exercise. American Journal ofPhysiology 276, C120–C127.

Balon, T.W., Zorzano, A., Treadway, J.L.,Goodman, M.N. & Ruderman, N.B.(1990) Effect of insulin on proteinsynthesis and degradation in skeletalmuscle after exercise. American Journal of Physiology 258, E92–E97.

Barger, P.M., Browning, A.C., Garner, A.N. & Kelly, D.P. (2001) p38 mitogen-activated protein kinase activatesperoxisome proliferator-activatedreceptor α. A potential role in the cardiac metabolic stress response.Journal of Biological Chemistry 276, 44 495–44 501.

Page 419: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 403

Bergstrom, J. & Hultman, E. (1966) Muscleglycogen synthesis after exercise: anenhancing factor localized to the musclecells in man. Nature 210, 309–310.

Bergstrom, J., Hultman, E. & Roch-Norlund, A.E. (1972) Muscle glycogensynthetase in normal subjects. Basalvalues, effect of glycogen depletion byexercise and of a carbohydrate-rich dietfollowing exercise. Scandinavian Journalof Clinical Laboratory Investigation 29,231–236.

Beyer, A., Kitzerow, A., Crute, B. et al.(2000) Muscle phosphorylase kinase isnot a substrate of AMP-activated proteinkinase. Biological Chemistry 381, 457–461.

Bjorntorp, P., Fahlen, M., Grimby, G. et al. (1972) Carbohydrate and lipidmetabolism in middle-aged physicallywell trained men. Metabolism 21,1037–1044.

Black, P.R., Brooks, D.C., Bessey, P.Q.,Wolfe, R.R. & Wilmore, D.W. (1982)Mechanisms of insulin resistancefollowing injury. Annals of Surgery 196,420–435.

Bogardus, C., Thuillex, P., Ravussin, E. et al. (1983) Effect of muscle glycogendepletion on in vivo insulin action inman. Journal of Clinical Investigation 72,1605–1610.

Bolster, D.R., Crozier, S.J., Kimball, S.R. & Jefferson, L.S. (2002) AMP-activatedprotein kinase suppresses proteinsynthesis in rat skeletal muscle throughdown-regulated mammalian target ofrapamycin (mTOR) signaling. Journal ofBiological Chemistry 277, 23 977–23 980.

Bonen, A., Tan, M.H. & Watson-Wright,W.M. (1984) Effects of exercise on insulinbinding and glucose metabolism inmuscle. Canadian Journal of Physiologyand Pharmacology 62, 1500–1504.

Booth, F.W. & Thomason, R.B. (1991)Molecular and cellular adaptation inresponse to exercise: perspectives ofvarious models. Physiology Reviews 71,541–585.

Boppart, M.D., Aronson, D., Gibson, L. et al. (1999) Eccentric exercise markedlyincreases c-Jun NH(2)-terminal kinaseactivity in human skeletal muscle.Journal of Applied Physiology 87,1668–1673.

Boppart, M.D., Asp, S., Wojtaszewski, J.F.et al. (2000) Marathon runningtransiently increases c-Jun NH2-terminal kinase and p38 activities inhuman skeletal muscle. Journal ofPhysiology 526, 663–669.

Boppart, M.D., Hirshman, M.F., Sakamoto,K., Fielding, R.A. & Goodyear, L.J.

inhibits glycogen synthase kinase-3 and activates glycogen synthase byrapamycin-insensitive pathways inskeletal muscle and adipose tissue. FEBSLetters 406, 211–215.

Dela, F., Ploug, T., Handberg, A. et al.(1994) Physical training increases muscleGLUT4 protein and mRNA in patientswith NIDDM. Diabetes 43, 862–865.

Dent, P., Lavoinne, A., Nakielny, S. et al.(1990) The molecular mechanism bywhich insulin stimulates glycogensynthesis in mammalian skeletal muscle.Nature 348, 302–308.

Devlin, J.T. & Horton, E.S. (1985) Effects ofprior high-intensity exercise on glucosemetabolism in normal and insulin-resistant men. Diabetes 34, 973–979.

Devlin, J.T., Hirshman, M.F., Horton, E.S.& Horton, E.D. (1987) Enhancedperipheral and splanchnic insulinsensitivity in NIDDM men after singlebout of exercise. Diabetes 36, 434–439.

Dohm, G.L., Kasperek, G.J., Tapscott, E.B.& Beecher, G.R. (1980) Effect of exerciseon synthesis and degradation of muscleprotein. Biochemical Journal 188, 255–262.

Donsmark, M., Langfort, J., Holm, C.,Ploug, T. & Galbo, H. (2003)Contractions activate hormone-sensitivelipase in rat muscle by protein kinase Cand mitogen-activated protein kinase.Journal of Physiology 550, 845–854.

Dufresne, S.D., Bjorbaek, C., El Haschimi,K. et al. (2001) Altered extracellularsignal-regulated kinase signaling and glycogen metabolism in skeletalmuscle from p90 ribosomal S6 kinase 2knockout mice. Molecular and CellularBiology 21, 81–87.

Elbrink, J. & Phipps, B.A. (1980) Studies on the persistence of enhancedmonosaccharide transport in rat skeletalmuscle following the cessation of theinitial stimulus. Cell Calcium 1, 349–358.

Farrell, P.A., Fedele, M.J., Vary, T.C. et al.(1999) Regulation of protein synthesisafter acute resistance exercise in diabeticrats. American Journal of Physiology 276,E721–E727.

Farrell, P.A., Hernandez, J.M., Fedele, M.J.et al. (2000) Eukaryotic initiation factorsand protein synthesis after resistanceexercise in rats. Journal of AppliedPhysiology 88, 1036–1042.

Fedele, M.J., Hernandez, J.M., Lang, C.H.et al. (2000) Severe diabetes prohibitselevations in muscle protein synthesisafter acute resistance exercise in rats.Journal of Applied Physiology 88, 102–108.

Fell, R.D., Terblanche, S.E., Ivy, J.L.,Young, J.C. & Holloszy, J.O. (1982) Effect

(2001) Static stretch increases c-JunNH2-terminal kinase activity and p38phosphorylation in rat skeletal muscle.American Journal of Physiology 280,C352–C358.

Brady, M.J., Bourbonais, F.J. & Saltiel, A.R. (1998) The activation of glycogensynthase by insulin switches from kinaseinhibition to phosphatase activationduring adipogenesis in 3T3-L1 cells.Journal of Biological Chemistry 273, 14 063–14 066.

Brozinick, J.T., Jr. & Birnbaum, M.J. (1998)Insulin, but not contraction, activatesAkt/PKB in isolated rat skeletal muscle.Journal of Biological Chemistry 273,14679–14682.

Buhl, E.S., Jessen, N., Schmitz, O. et al.(2001) Chronic treatment with 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside increases insulin-stimulated glucose uptake and GLUT4translocation in rat skeletal muscles in afiber type-specific manner. Diabetes 50,12–17.

Burstein, R., Polychronakos, C., Toews, C.J. et al. (1985) Acute reversal of theenhanced insulin action in trainedathletes. Diabetes 34, 756–760.

Carling, D. & Hardie, D.G. (1989) Thesubstrate and sequence specificity of the AMP-activated protein kinase.Phosphorylation of glycogen synthaseand phosphorylase kinase. Biochimica etBiophysica Acta 1012, 81–86.

Carlson, C.J., Fan, Z., Gordon, S.E. &Booth, F.W. (2001) Time course of theMAPK and PI3-kinase response within24 h of skeletal muscle overload. Journalof Applied Physiology 91, 2079–2087.

Chibalin, A.V., Yu, M., Ryder, J.W. et al.(2000) Exercise-induced changes inexpression and activity of proteinsinvolved in insulin signal transductionin skeletal muscle: differential effects oninsulin-receptor substrates 1 and 2.Proceedings of the National Academy ofSciences of the United States of America 97,38–43.

Conlee, R.K., Hickson, R.C., Winder,W.W., Hagberg, J.M. & Holloszy, J.O.(1978) Regulation of glycogenresynthesis in muscles of rats followingexercise. American Journal of Physiology235, R145–R150.

Cross, D.A.E., Alessi, D.R., Cohen, P.,Andjelkovich, M. & Hemmings, B.A.(1995) Inhibition of glycogen synthasekinase-3 by insulin mediated by proteinkinase B. Nature 378, 785–789.

Cross, D.A.E., Watt, P.W., Shaw, M. et al.(1997) Insulin activates protein kinase B,

Page 420: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

404 chapter 27

of muscle glycogen content on glucoseuptake following exercise. Journal ofApplied Physiology 52, 434–437.

Fluckey, J.D., Vary, T.C., Jefferson, L.S. & Farrell, P.A. (1996) Augmented insulin action on rates of proteinsynthesis after resistance exercise in rats. American Journal of Physiology 270,E313–E319.

Fluckey, J.D., Ploug, T. & Galbo, H. (1999)Attenuated insulin action on glucoseuptake and transport in musclefollowing resistance exercise in rats. Acta Physiologica Scandinavica 167, 77–82.

Force, T. & Bonventre, J.V. (1998) Growthfactors and mitogen-activated proteinkinases. Hypertension 31, 152–161.

Fujii, N., Boppart, M.D., Dufresne, S.D. et al. (2001) Overexpression of JNK inskeletal muscle does not alter glycogensynthase activity. Diabetes 50, A276.

Garcia-Roves, P.M., Han, D.H., Song, Z. et al. (2003) Prevention of glycogensupercompensation prolongs theincrease in muscle GLUT4 after exercise.American Journal of Physiology 285,E729–E736.

Gomez, D.A., Martinez-Martinez, S.,Maldonado, J.L., Ortega-Perez, I. &Redondo, J.M. (2000) A role for the p38MAP kinase pathway in the nuclearshuttling of NFATp. Journal of BiologicalChemistry 275, 13 872–13 878.

Goodyear, L.J., Giorgino, F., Balon, T.W.,Condorelli, G. & Smith, R.J. (1995)Effects of contractile activity on tyrosinephophoproteins and PI 3-kinase activityin rat skeletal muscle. American Journal ofPhysiology 268, E987–E995.

Goodyear, L.J., Chang, P.-Y., Sherwood,D.J., Dufresne, S.D. & Moller, D.E. (1996) Effects of exercise and insulin on mitogen-activated protein kinasesignaling pathways in rat skeletalmuscle. American Journal of Physiology271, E403–E408.

Greiwe, J.S., Hickner, R.C., Hansen, P.A. et al. (1999) Effects of endurance exercise training on muscle glycogenaccumulation in humans. Journal ofApplied Physiology 87, 222–226.

Haddad, F. & Adams, G.R. (2002) Selectedcontribution: acute cellular andmolecular responses to resistanceexercise. Journal of Applied Physiology 93,394–403.

Hamilton, S.R., Stapleton, D., O’Donnell,J.B. et al. (2001) An activating mutationin the γ1 subunit of the AMP-activatedprotein kinase. FEBS Letters 500,163–168.

Hollenbeck, C.B., Haskell, W., Rosenthal,M. & Reaven, G.M. (1984) Effect ofhabitual physical activity on regulationof insulin-stimulated glucose disposal inolder males. Journal of the AmericanGeriatric Society 33, 273–277.

Holloszy, J.O. & Narahara, H.T. (1965) Changes in permeability to 3-methylglucose associated withcontraction of isolated frog muscle.Journal of Biological Chemistry 240,3493–3500.

Horman, S., Browne, G., Krause, U. et al. (2002) Activation of AMP-activated protein kinase leads to thephosphorylation of elongation factor 2and an inhibition of protein synthesis.Current Biology 12, 1419–1423.

Host, H.H., Hansen, P.A., Nolte, L.A.,Chen, M.M. & Holloszy, J.O. (1998)Glycogen supercompensation masks the effect of a traininginduced increasein GLUT-4 on muscle glucose transport.Journal of Applied Physiology 85, 133–138.

Houmard, J.A., Shaw, C.D., Hickey, M.S. & Tanner, C.J. (1999) Effect of short-termexercise training on insulin-stimulatedPI 3-kinase activity in human skeletalmuscle. American Journal of Physiology277, E1055–E1060.

Howlett, K.F., Sakamoto, K., Hirshman,M.F. et al. (2002) Insulin signaling afterexercise in insulin receptor substrate-2-deficient mice. Diabetes 51, 479–483.

Hughes, V.A., Fiatarone, M.A., Fielding,R.A. et al. (1993) Exercise increasesmuscle GLUT-4 levels and insulin actionin subjects with impaired glucosetolerance. American Journal of Physiology264, E855–E862.

Ihlemann, J., Ploug, T., Hellsten, Y. &Galbo, H. (1999) Effect of tension oncontraction-induced glucose transport in rat skeletal muscle. American Journal ofPhysiology 277, E208–E214.

Ivy, J.L. & Holloszy, J.O. (1981) Persistentincrease in glucose uptake by rat skeletalmuscle following exercise. AmericanJournal of Physiology 241, C200–C203.

Johansen, K. & Munch, O. (1979) Therelationship between maximal oxygenuptake and glucose tolerance/insulinresponse ratio in normal young men.Hormone and Metabolic Research 11,424–427.

Jorgensen, S.B., Viollet, B., Andreelli, F. et al. (2004) Knockout of the α2 but not α1 5′-AMP-activated protein kinaseisoform abolishes 5-aminoimidazole-4-carboxamide-1-β-4-ribofuranosidebutnot contraction-induced glucose uptake

Han, J., Jiang, Y., Li, Z., Kravchenko, V.V.& Ulevitch, R.J. (1997) Activation of thetranscription factor MEF2C by the MAPkinase p38 in inflammation. Nature 386,296–299.

Hardie, D.G., Carling, D. & Carlson, M.(1998) The AMP-activated/SNF1protein kinase subfamily: metabolicsensors of the eukaryotic cell. AnnualReviews of Biochemistry 67, 821–855.

Haruta, T., Morris, A.J., Rose, D.W. et al.(1995) Insulin-stimulated GLUT4translocation is mediated by a divergentintracellular signaling pathway. Journal of Biological Chemistry 270, 27 991–27 994.

Hawley, S.A., Boudeau, J., Reid, J.L. et al.(2003) Complexes between the LKB1tumor suppressor, STRADα/β andMO25α/β are upstream kinases in theAMP-activated protein kinase cascade.Journal of Biology 2, 28.1–28.16.

Hayashi, T., Hirshman, M.F., Kurth, E.J.,Winder, W.W. & Goodyear, L.J. (1998)Evidence for 5′ AMP-activated proteinkinase mediation of the effect of musclecontraction on glucose transport.Diabetes 47, 1369–1373.

Hayashi, T., Hirshman, M.F., Dufresne,S.D. & Goodyear, L.J. (1999) Skeletalmuscle contractile activity in vitrostimulates mitogen-activated proteinkinase signaling. American Journal ofPhysiology 277, C701–C707.

Hayashi, T., Hirshman, M.F., Fujii, N. et al. (2000) Metabolic stress and alteredglucose transport: Activation of AMP-activated protein kinase as a unifyingcoupling mechanism. Diabetes 49,527–531.

Hazzalin, C.A. & Mahadevan, L.C. (2002)MAPK-regulated transcription: Acontinuously variable gene switch.Molecular and Cellular Biology 3, 30–40.

Heath, G.W., Gavin I.I., Hinderliter, J.M. et al. (1983) Effects of exercise and lack ofexercise on glucose tolerance and insulinsensitivity. Journal of Applied Physiology55, 628–634.

Helmrich, S.P., Ragland, D.R., Leung, R.W.& Paffenbarger, R.S. (1991) Physicalactivity and reduced occurrence of non-insulin-dependant diabetes mellitus.New England Journal of Medicine 325,147–152.

Ho, R.C., Alcazar, O., Fujii, N., Hirshman,M.F. & Goodyear, L.J. (2004) p38 γMAPK regulation of glucose transporterexpression and glucose uptake in L6myotubes and mouse skeletal muscle.American Journal of Physiology 286,R342–R349.

Page 421: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 405

in skeletal muscle. Journal of BiologicalChemistry 279, 1070–1079.

Kawanaka, K., Nolte, L.A., Han, D.H.,Hansen, P.A. & Holloszy, J.O. (2000)Mechanisms underlying impairedGLUT-4 translocation in glycogen-supercompensated muscles of exercisedrats. American Journal of Physiology 279,E1311–E1318.

Kimball, S.R., Farrell, P.A. & Jefferson, L.S.(2002) Invited review: role of insulin intranslational control of protein synthesisin skeletal muscle by amino acids orexercise. Journal of Applied Physiology 93,1168–1180.

Kimura, N., Tokunaga, C., Dalal, S. et al.(2003) A possible linkage between AMP-activated protein kinase (AMPK)and mammalian target of rapamycin(mTOR) signalling pathway. Genes toCells 8, 65–79.

King, D.S., Dalsky, G.P., Staten, M.A. et al.(1987) Insulin action and secretion inendurance trained and untrainedhumans. Journal of Applied Physiology 63,2247–2252.

King, D.S., Dalsky, G.P., Cutler, W.E. et al. (1988) Effects of lack of exercise oninsulin secretion and action in trainedsubjects. American Journal of Physiology254, E537–E542.

Kirwan, J.P., Hickner, R.C., Yarasheski,K.E. et al. (1992) Eccentric exerciseinduces transient insulin resistance inhealthy individuals. Journal of AppliedPhysiology 72, 2197–2202.

Kirwan, J.P., Del Aguila, L.F., Hernandez,J.M. et al. (2000) Regular exerciseenhances insulin activation of IRS-1-associated PI3-kinase in human skeletalmuscle. Journal of Applied Physiology 88,797–803.

Knutti, D., Kressler, D. & Kralli, A. (2001)Regulation of the transcriptionalcoactivator PGC-1 via MAPK-sensitiveinteraction with a repressor. Proceedingsof the National Academy of Sciences of theUnited States of America 98, 9713–9718.

Kolterman, O.G., Insel, J., Seakow, M. &Olefsky, J.M. (1980) Mechanisms ofinsulin resistance in human obesity:evidence for receptor and post-receptordefects. Journal of Clinical Investigation 65,1272–1284.

Kostyak, J.C., Kimball, S.R., Jefferson, L.S.& Farrell, P.A. (2001) Severe diabetesinhibits resistance exercise-inducedincrease in eukaryotic initiation factor 2Bactivity. Journal of Applied Physiology 91,79–84.

Krause, U., Bertrand, L. & Hue, L. (2002)Control of p70 ribosomal protein S6

kinase/Akt-1 pathway. European Journalof Endocrinology 147, 149–157.

Lund, S., Pryor, P.R., Ostergaard, S. et al.(1998) Evidence against protein kinase B as a mediator of contraction-inducedglucose transport and GLUT4translocation in rat skeletal muscle.FEBS Letters 425, 472–474.

Manson, J.E., Rimm, E.B., Stampfer, M.J. et al. (1991) Physical activity andincidence of non-insulin dependentdiabetes mellitus in women. Lancet 338,774–778.

Manson, J.E., Nathan, D.M., Krolewski,A.S. et al. (1992) A prospective study of exercise and incidence of diabetesamong US male physicians. Journal of theAmerican Medical Association 268, 63–67.

Markuns, J.F., Wojtaszewski, J.F.P. &Goodyear, L.J. (1999) Insulin andexercise decrease glycogen synthasekinase-3 activity by differentmechanisms in rat skeletal muscle.Journal of Biological Chemistry 274, 24 896–24 900.

Martineau, L.C. & Gardiner, P.F. (2001) Insight into skeletal musclemechanotransduction: MAPK activationis quantitatively related to tension.Journal of Applied Physiology 91, 693–702.

Merrall, N.W., Plevin, R.J., Stokoe, D. et al.(1993) Mitogen-activated protein kinase(MAP kinase), MAP kinase kinase and c-Mos stimulate glucose transport inXenopus oocytes. Biochemical Journal 295,351–355.

Mikines, K.J., Sonne, B., Farrell, P.A.,Tronier, B. & Galbo, H. (1988) Effect ofphysical exercise on sensitivity andresponsiveness to insulin in humans.American Journal of Physiology 254,E248–E259.

Mikines, K.J., Sonne, B., Farrell, P.A.,Tronier, B. & Galbo, H. (1989) Effect of training on the dose–responserelationship for insulin action in men.Journal of Applied Physiology 66, 695–703.

Milan, D., Jeon, J.T., Looft, C. et al. (2000) A mutation in PRKAG3 associated withexcess glycogen content in pig skeletalmuscle. Science 288, 1248–1251.

Miller, J.P., Pratley, R.E., Goldberg, A.P. et al. (1994) Strength training increasesinsulin action in healthy 50- to 65-yr-oldmen. Journal of Applied Physiology 77,1122–1127.

Moxham, C.M., Tabrizchi, A., Davis, R.J. & Malbon, C.C. (1996) Jun N-terminalkinase mediates activation of skeletalmuscle glycogen synthase by insulin in

kinase and acetyl-CoA carboxylase byAMP-activated protein kinase andprotein phosphatases in isolatedhepatocytes. European Journal ofBiochemistry 269, 3751–3759.

Kristiansen, S., Asp, S. & Richter, E.A.(1996) Decreased muscle GLUT-4 andcontraction-induced glucose transportafter eccentric contractions. AmericanJournal of Physiology 271, R477–R482.

Krook, A., Widegren, U., Jiang, X.J. et al.(2000) Effects of exercise on mitogenactivated protein kinase. AmericanJournal of Physiology 279, R1716–R1721.

Kyriakis, J.M. & Avruch, J. (2001)Mammalian mitogen-activated proteinkinase signal transduction pathwaysactivated by stress and inflammation.Physiology Reviews 81, 807–869.

Langfort, J., Donsmark, M., Ploug, T.,Holm, C. & Galbo, H. (2003) Hormone-sensitive lipase in skeletal muscle:regulatory mechanisms. ActaPhysiologica Scandinavica 178, 397–403.

Lazar, D.F., Wiese, R.J., Brady, M.J. et al.(1995) Mitogen-activated protein kinasekinase inhibition does not block thestimulation of glucose utilization byinsulin. Journal of Biological Chemistry270, 20 801–20 807.

LeBlanc, J., Nadeau, A., Boulay, M. &Rousseau-Migneron, S. (1979) Effects of physical training and adiposity onglucose metabolism and 125I-insulinbinding. Journal of Applied Physiology 46,235–239.

LeBlanc, J., Nadeau, A., Richard, D. &Tremblay, A. (1981) Studies on thesparing effect of exercise on insulinrequirements in human subjects.Metabolism 30, 1119–1124.

Liu, J. & Brautigan, D.L. (2000) Insulin-stimulated phosphorylation of theprotein phosphatase-1 striated muscleglycogen-targeting subunit andactivation of glycogen synthase. Journalof Biological Chemistry 275, 15 940–15 947.

Lizcano, J.M., Goransson, O., Toth, R. et al.(2004) LKB1 is a master kinase thatactivates 13 kinases of the AMPKsubfamily, including MARK/PAR-1.EMBO Journal 23, 833–843.

Lohman, D., Liebold, F., Heilmann, W.,Singer, J. & Pohl, A. (1978) Diminishedinsulin response in highly trainedathletes. Metabolism Clinical andExperimental 27, 521–524.

Luciano, E., Carneiro, E.M., Carvalho, C.R. et al. (2002) Endurance trainingimproves responsiveness to insulin andmodulates insulin signal transductionthrough the phosphatidylinositol 3-

Page 422: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

406 chapter 27

vivo. Journal of Biological Chemistry 271, 30 765–30 773.

Mu, J., Brozinick, J.T., Jr., Valladares, O.,Bucan, M. & Birnbaum, M.J. (2001) Arole for AMP-activated protein kinase in contraction- and hypoxia-regulatedglucose transport in skeletal muscle.Molecular Cell 7, 1085–1094.

Nader, G.A. & Esser, K.A. (2001)Intracellular signaling specificity inskeletal muscle in response to differentmodes of exercise. Journal of AppliedPhysiology 90, 1936–1942.

Nakatani, A., Han, D.H., Hansen, P.A. et al. (1997) Effect of endurance exercisetraining on muscle glycogensupercompensation in rats. Journal ofApplied Physiology 82, 711–715.

Oshida, Y., Yamanouchi, K., Hayamizu, S.& Sato, Y. (1989) Long-term mild joggingincreases insulin action despite noinfluence on body mass index or Vo2max.Journal of Applied Physiology 66,2206–2210.

Osman, A.A., Pendergrass, M., Koval, J. et al. (2000) Regulation of MAP kinasepathway activity in vivo in humanskeletal muscle. American Journal ofPhysiology 278, E992–E999.

Ossipova, O., Bardeesy, N., DePinho, R.A. & Green, J.B. (2003) LKB1 (XEEK1)regulates Wnt signalling in vertebratedevelopment. Nature Cell Biology 5,889–894.

Price, T.B., Laurent, D. & Petersen, K.F.(2003) 13C/31P NMR studies on the roleof glucose transport/phosphorylation in human glycogen supercompensation.International Journal of Sports Medicine 24,238–244.

Pruett, E.D.R. & Oseid, S. (1970) Effect ofexercise on glucose and insulin responseto glucose infusion. Scandinavian Journalof Clinical and Laboratory Investigation 26,277–285.

Puigserver, P. & Spiegelman, B.M. (2003)Peroxisome proliferator-activatedreceptor-γ coactivator 1α (PGC-1α):transcriptional coactivator andmetabolic regulator. EndocrinologyReviews 24, 78–90.

Puigserver, P., Rhee, J., Lin, J. et al. (2001)Cytokine stimulation of energyexpenditure through p38 MAP kinaseactivation of PPARγ coactivator-1.Molecular Cell 8, 971–982.

Rasmussen, B.B. & Winder, W.W. (1997)Effect of exercise intensity on skeletalmuscle malonyl-CoA and acetyl-CoAcarboxylase. Journal of Applied Physiology83, 1104–1109.

Richter, E.A., Garetto, L.P., Goodman,

GLUT4 translocation. Journal of BiologicalChemistry 278, 14 599–14 602.

Sasson, S., Edelson, D. & Cerasi, E. (1987)In vitro autoregulation of glucoseutilization in rat soleus muscle. Diabetes36, 1041–1046.

Seals, D.R., Hagberg, J.M., Allen, W.K. et al. (1984) Glucose tolerance in youngand older athletes and sedentary men.Journal of Applied Physiology 56,1521–1525.

Shah, O.J., Anthony, J.C., Kimball, S.R. &Jefferson, L.S. (2000) 4E-BP1 and S6K1:translational integration sites fornutritional and hormonal information in muscle. American Journal of Physiology279, E715–E729.

Sherwood, D.J., Dufresne, S.D., Markuns,J.F. et al. (1999) Differential regulation ofMAP kinase, p70 S6 kinase and Akt bycontraction and insulin in rat skeletalmuscle. American Journal of Physiology276, E870–E878.

Skurat, A.V., Wang, Y. & Roach, P.J. (1994)Rabbit skeletal muscle glycogensynthase expressed in COS cells.Identification of regulatoryphosphorylation sites. Journal ofBiological Chemistry 269, 25 534–25 542.

Somwar, R., Perreault, M., Kapur, S. et al.(2000) Activation of p38 mitogen-activated protein kinase α and by insulinand contraction in rat skeletal muscle:potential role in the stimulation ofglucose transport. Diabetes 49,1794–1800.

Sutherland, C., Leighton, I.A. & Cohen, P.(1993) Inactivation of glycogen synthasekinase-3β by phosphorylation: newkinase connections in insulin andgrowth-factor signalling. BiochemicalJournal 296, 15–19.

Suzuki, Y., Lanner, C., Kim, J.H. et al.(2001) Insulin control of glycogenmetabolism in knockout mice lackingthe muscle-specific protein phosphatasePP1G/RGL. Molecular and Cellular Biology21, 2683–2694.

Sweeney, G., Somwar, R., Ramlal, T. et al.(1999) An inhibitor of p38 mitogen-activated protein kinase preventsinsulin-stimulated glucose transport butnot glucose transporter translocation in3T3-L1 adipocytes and L6 myotubes.Journal of Biological Chemistry 274, 10 071–10 078.

Tanner, C.J., Koves, T.R., Cortright, R.L. et al. (2002) Effect of short-term exercisetraining on insulin-stimulated PI 3-kinase activity in middle-aged men.American Journal of Physiology 282,E147–E153.

M.N. & Ruderman, N.B. (1982) Muscleglucose metabolism following exercisein the rat: increased sensitivity toinsulin. Journal of Clinical Investigation 69,785–793.

Richter, E.A., Garreto, L.P., Goodman,M.N., Ruderman, N.B. & Garetto, L.P.(1984) Enhanced muscle glucosemetabolism after exercise: modulationby local factors. American Journal ofPhysiology 246, E476–E482.

Richter, E.A., Mikines, K.J., Galbo, H. &Kiens, B. (1989) Effect of exercise oninsulin action in human skeletal muscle.Journal of Applied Physiology 66, 876–885.

Rosenthal, M., Haskell, W.L., Solomon, R.,Widstrom, A. & Reaven, G.M. (1983)Demonstration of a relationshipbetween level of physical training andinsulin-stimulated glucose utilization in normal humans. Diabetes 32, 408–411.

Ruderman, N.B., Houghton, C.R.S. &Hems, R. (1971) Evaluation of theisolated perfused rat hindquarter for thestudy of muscle metabolism. BiochemicalJournal 124, 639–651.

Russell, R.R., III, Bergeron, R., Shulman,G.I. & Young, L.H. (1999) Translocationof myocardial GLUT-4 and increasedglucose uptake through activation ofAMPK by AICAR. American Journal ofPhysiology 277, H643–H649.

Ryder, J.W., Fahlman, R., Wallberg-Henriksson, H. et al. (2000) Effect ofcontraction on mitogen-activatedprotein kinase signal transduction inskeletal muscle. Journal of BiologicalChemistry 275, 1457–1462.

Sakamoto, K. & Goodyear, L.J. (2002)Invited review: intracellular signaling in contracting skeletal muscle. Journal ofApplied Physiology 93, 369–383.

Sakamoto, K., Hirshman, M.F.,Aschenbach, W.G. & Goodyear, L.J.(2002) Contraction regulation of Akt inrat skeletal muscle. Journal of BiologicalChemistry 277, 11 910–11 917.

Sakamoto, K., Aschenbach, W.G.,Hirshman, M.F. & Goodyear, L.J. (2003)Akt signaling in skeletal muscle:regulation by exercise and passivestretch. American Journal of Physiology285, E1081–E1088.

Saltin, G., Lingard, F., Houston, M. et al.(1978) Physical training and glucosetolerance in middle aged men withchemical diabetes. Diabetes 28, 30–32.

Sano, H., Kane, S., Sano, E. et al. (2003)Insulin-stimulated phosphorylation of aRab GTPase-activating protein regulates

Page 423: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

exercise regulation of insulin action 407

Tanti, J.F., Gremeaux, T., Grillo, S. et al.(1996) Overexpression of a constitutivelyactive form of phosphatidylinositol 3-kinase is sufficient to promote Glut 4translocation in adipocytes. Journal ofBiological Chemistry 271, 25 227–25 232.

Tipton, K.D. & Wolfe, R.R. (2001) Exercise,protein metabolism, and muscle growth.International Journal of Sport NutritionExercise and Metabolism 11, 109–132.

Todd, M.K. & Turcotte, L. (2003) ERKinhibition abolishes the contraction-induced increase in muscle FA uptake.FASEB Journal 17, 317.7 [abstract].

Treadway, J.L., James, D.E., Burcel, E. &Ruderman, N.B. (1989) Effect of exerciseon insulin receptor binding and kinaseactivity in skeletal muscle. AmericanJournal of Physiology 256, E138–E144.

Trovati, M., Carta, Q., Cavalot, F. et al.(1984) Influence of physical training onblood glucose control, glucose tolerance,insulin secretion, and insulin action innon-insulin-dependent diabeticpatients. Diabetes Care 7, 416–420.

Turinsky, J. & Damrau-Abney, A. (1999)Akt kinases and 2-deoxyglucose uptakein rat skeletal muscles in vivo: study withinsulin and exercise. American Journal ofPhysiology 276, R277–R282.

Vavvas, D., Apazidis, A., Saha, A.K. et al.(1997) Contraction-induced changes inacetyl-CoA carboxylase and 5′-AMP-activated kinase in skeletal muscle.Journal of Biological Chemistry 272, 13 255–13 261.

Wallberg-Henriksson, H. & Holloszy, J.O.(1984) Contractile activity increasesglucose uptake by muscle in severelydiabetic rats. Journal of Applied Physiology57(4), 1045–1049.

Wallberg-Henriksson, H. & Holloszy, J.O.(1985) Activation of glucose transport in diabetic muscle: responses tocontraction and insulin. American Journalof Physiology 249, C233–C237.

Wardzala, L.J., Hirshman, M.F., Pofcher, E.et al. (1985) Regulation of glucoseutilization in adipose cells and muscleafter long-term experimentalhyperinsulinemia in rats. Journal ofClinical Investigation 76, 460–469.

Watt, M.J., Stellingwerff, T., Heigenhauser,G.J. & Spriet, L.L. (2003) Effects ofplasma adrenaline on hormone-sensitivelipase at rest and during moderateexercise in human skeletal muscle.Journal of Physiology 550, 325–332.

Wojtaszewski, J.F.P., Nielsen, P., Kiens, B.& Richter, E.A. (2001) Regulation ofglycogen synthase kinase-3 in humanskeletal muscle: Effects of food intakeand bicycle exercise. Diabetes 50,265–269.

Wojtaszewski, J.F.P., Jorgensen, S.B.,Hellsten, Y., Hardie, D.G. & Richter, E.A.(2002) Glycogen-dependent effects of 5-aminoimidazole-4-carboxamide(AICA) -riboside on AMP-activatedprotein kinase and glycogen synthaseactivities in rat skeletal muscle. Diabetes51, 284–292.

Wretman, C., Widegren, U., Lionikas, A.,Westerblad, H. & Henriksson, J. (2000)Differential activation of mitogen-activated protein kinase signallingpathways by isometric contractions inisolated. Acta Physiologica Scandinavica170, 45–49.

Wretman, C., Lionikas, A., Widegren, U.et al. (2001) Effects of concentric and eccentric contractions onphosphorylation of MAPK(erk1/2) and MAPK(p38) in isolated rat skeletalmuscle. Journal of Physiology 535,155–164.

Yaspelkis, B.B., III, Singh, M.K., Trevino,B., Krisan, A.D. & Collins, D.E. (2002)Resistance training increases glucoseuptake and transport in rat skeletalmuscle. Acta Physiologica Scandinavica175, 315–323.

Young, J.C., Garthwaite, S.M., Bryan, J.E. et al. (1983) Carbohydrate feeding speedsreversal of enhanced glucose uptake inmuscle after exercise. American Journal ofPhysiology 245, R684–R688.

Yu, M., Blomstrand, E., Chibalin, A.V.,Krook, A. & Zierath, J.R. (2001)Marathon running increases ERK1/2and p38 MAP kinase signalling todownstream targets in human skeletalmuscle. Journal of Physiology 536,273–282.

Zorzano, A., Balon, T.W., Garetto, L.P.,Goodman, M.N. & Ruderman, N.B.(1985) Muscle α-aminoisobutyric acidtransport after exercise: enhancedstimulation by insulin. American Journalof Physiology 248, 546–552.

Zorzano, A., Balon, T.W., Goodman, M.N.& Ruderman, N.B. (1986) Glycogendepletion and increased insulinsensitivity and responsiveness in muscleafter exercise. American Journal ofPhysiology 251, E664–E669.

Welsh, G.I., Miller, C.M., Loughlin, A.J.,Price, N.T. & Proud, C.G. (1998)Regulation of eukaryotic initiation factor eIF2B: glycogen synthase kinase-3phosphorylates a conserved serinewhich undergoes dephosphorylation in response to insulin. FEBS Letters 421,125–130.

Widegren, U., Jiang, X.J., Krook, A. et al.(1998) Divergent effects of exercise onmetabolic and mitogenic signalingpathways in human skeletal muscle.FASEB Journal 12, 1379–1389.

Widegren, U., Ryder, J.W. & Zierath, J.R.(2001) Mitogen-activated protein kinasesignal transduction in skeletal muscle:effects of exercise and musclecontraction. Acta PhysiologicaScandinavica 172, 227–238.

Winder, W.W. & Hardie, D.G. (1996)Inactivation of acetyl-CoA carboxylaseand activation of AMP-activated proteinkinase in muscle during exercise.American Journal of Physiology 270,E299–E304.

Winder, W.W., Holmes, B.F., Rubink, D.S.et al. (2000) Activation of AMP-activatedprotein kinase increases mitochondrialenzymes in skeletal muscle. Journal ofApplied Physiology 88, 2219–2226.

Wojtaszewski, J.F.P., Hansen, B.F., Urso, B.& Richter, E.A. (1996) Wortmannininhibits both insulin- and contraction-stimulated glucose uptake and transportin rat skeletal muscle. Journal of AppliedPhysiology 81, 1501–1509.

Wojtaszewski, J.F.P., Hansen, B.F., Kiens, B. & Richter, E.A. (1997) Insulinsignaling in human skeletal muscle: timecourse and effect of exercise. Diabetes 46,1775–1781.

Wojtaszewski, J.F.P., Higaki, Y., Hirshman,M.F. et al. (1999a) Exercise modulatespostreceptor insulin signaling andglucose transport in muscle-specificinsulin receptor knockout mice. Journalof Clinical Investigation 104, 1257–1264.

Wojtaszewski, J.F.P., Lynge, J., Jakobsen,A.B., Goodyear, L.J. & Richter, E.A.(1999b) Differential regulation of MAPkinase by contraction and insulin inskeletal muscle: metabolic implications.American Journal of Physiology 277,E724–E732.

Wojtaszewski, J.F.P., Hansen, B.F., Gade, J.et al. (2000) Insulin signaling and insulinsensitivity after exercise in humanskeletal muscle. Diabetes 49, 325–331.

Page 424: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

408

Introduction

Physical activity is an essential component of normal well-being. Within the last decade, moreindividuals have undertaken activity programs inorder to ameliorate chronic medical conditions or to maintain their healthy status than ever before.Some believe that long-term exercises will improvelongevity and quality of life, while others contendthat physical fitness enhances mental well-being as well as improving muscle function. Regardless of the physiology or the rationale, the long-termeffects, either beneficial or harmful, associated withchronic exercise are an area of intense investigation.

By the nature of its components, physical activ-ity affects almost all hormonal systems, from β-endorphins in the brain, to local cytokines andchemokines in bone. Although the vast majority ofpeople believe the effects of long-term exercise arebeneficial to virtually every organ system, drasticchanges in the normal physiologic state of these hormonal modulators can impact tissues in a negat-ive as well as positive way. Nowhere is this moreapparent than in the clinical syndrome of exercise-induced amenorrhea and the female athlete’s triad. In predominantly younger women, chronicand intense physical activity results in loss of thegonadotropin pulse generator leading to estrogendeprivation. This in turn drastically affects the boneremodeling unit such that bone resorption exceedsformation resulting in significant bone loss. As such,this interface between homeostatic processes relatedto hormonal signaling and stress mechanisms in-duced by exercise are most noticeable in bone, an

organ infrequently thought of as a target tissue forsystemic modulators. Yet the list of skeletal mediatorsaffected by chronic exercise is nearly endless andincludes significant changes in circulating gonadalsteroids, adrenal steroids, cytokines, prostaglandins,growth hormone (GH), insulin, insulin-like growthfactor I (IGF-I), leptin and others. All these endo-genous compounds influence other systems such asmetabolic fuel balance, cardiovascular fitness andmuscle integrity to induce changes that may eitherbe beneficial or harmful to the organism.

Weightlessness with manned space flight and theaccompanying animal experiments provided thefirst in vivo proof of the importance of hormonaleffectors on skeletal growth, remodeling and mass.Since those early days of space exploration, ourunderstanding of the effects of physical training onhormonal balance, and its subsequent actions on theskeleton, has grown immensely. But there are stillmany unanswered questions. In this review, I willfocus first on the physiology of bone remodelingsince that provides the background for understand-ing skeletal homeostasis and the importance of cir-culating, as well as local growth factors in mediatingadaptive processes in the skeleton. Next, I will touchon the effects of acute exercise on bone cell function,with a particular focus on the cellular aspects ofstrain within the basic multicellular unit (BMU) ofbone. In the Hormonal and skeletal responses tolong-term exercise section below, I will elaborate onthe complex skeletal changes associated with sus-tained exercise programs, particularly as they relateto hormonal balances and imbalances. Finally, I willdiscuss some potential clinical implications of our

Chapter 28

Hormone and Exercise-Induced Modulation ofBone Metabolism

CLIFFORD J. ROSEN

Page 425: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 409

current knowledge about exercise, hormonal modu-lation and skeletal homeostasis.

Physiology of normal and aberrant bone remodeling

Normal modeling and remodeling

Mammalian skeletons grow and remodel duringlife. Linear growth is accomplished at the growthplate and is regulated by GH and IGF-I. In rodentslinear growth continues over a lifetime although it is most pronounced during puberty. On the otherhand, the human skeleton which is more than justcalcium phosphate crystals melded together in aprotein matrix, grows, models and then remodels.Linear growth in humans occurs from the growthplate, begins at birth and ceases after puberty. It isprincipally modulated by growth plate chondro-cytes. Modeling, which is essentially the process ofshaping a bone through resorption and formation,occurs in response to several factors includinghumoral substances, muscle activity and local factors.It too ceases after puberty in response to severalcues including changes in sex steroids and circulat-ing IGF-I. Modeling is very dependent on the direc-tion of the stress vectors that are modulated throughmuscle such that the actual cross-sectional shape ofbone is not perfectly oval but rather slightly eccen-tric, depending on forces shaping it by loading.Remodeling is a very distinct homeostatic processcompared to growth and modeling, even though the cellular players are similar. Remodeling allowsthe skeleton to reorganize itself without changing it absolute mass and thus serves to enhance skel-etal integrity while maintaining metabolic balance,especially for essential ions such as calcium andphosphate (Rosen 2003). During remodeling, therate of bone resorption or dissolution equals the rate of new bone formation. In contrast, new bone is added by growth and modeling due to linearexpansion from the growth plate by chondrocytes,and expansion of lateral surfaces in the diaphysis by periosteal osteoblasts (OBs). Acquisition of peakbone mass requires optimization of all three distinctbut overlapping processes. General physical activ-ity affects the growing skeleton, particularly at a

time when these three distinct activities are at theirhighest level; i.e. adolescence. From about the age of10–18 years, linear growth is very active and model-ing of the skeleton is in full force. As noted below,most studies that have examined the role of physicalactivity (i.e. loading the skeleton in one form oranother, such as running or weightlifting) on bonehave noted a much more vigorous response inrespect to changes in bone mineral density (BMD)during this time period than any other.

Remodeling is a constant process that dictates the metabolic needs of the skeleton and provides forthe elasticity necessary for general physical activity.Every 10 years in humans the entire skeleton isremodeled with the greatest turnover noted in thetrabecular rich regions of the thoraco-lumbar spineand several areas of the femur (Rosen 2003). Becauseof this huge undertaking, it is not surprising that the mammalian skeleton is a highly organized andphysiologically active organ. Bone basically servestwo purposes: (i) to maintain structure; and (ii) topreserve calcium homeostasis for all physiologicprocesses. As such, the mammalian skeleton isuniquely designed for its protective and structuralroles. There is an outer surface of cortical bone thatsurrounds the inner trabecular elements (Fig. 28.1).Marrow bathes the trabecular skeleton while cor-tical bone is nourished by periosteal vessels and a

Trabecular bone

Bone is a two component organ:cortical and trabecular bone analyzed by mCT

Cortical bone

Fig. 28.1 The two compartment model of the skeleton.The inner skeletal complex is made up of trabecularelements with a wide surface area that is bathed in bonemarrow. The outer shell is the cortex and on the far outersurface of the cortex is the perisoteal membrane. µCT,micro computed tomography.

Page 426: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

410 chapter 28

series of canaliculi connecting osteocytes to liningcells and OBs (Fig. 28.2). The BMU defines the singlefunctional component of bone remodeling andincludes lining cells, OBs, osteoclasts (OCs) andosteocytes (Figs 28.2 and 28.3). Gravitary forcesinfluence the BMU and stimulate cortical and tra-becular remodeling. In respect to bone growth,periosteal OBs and the underlying growth plate areprincipally responsible for longitudinal growth andlateral expansion. Both cortical and trabecular boneundergo remodeling, but the frequency of this pro-cess is much less in the cortex than in the trabecularcomponents of the spine and distal femur.

Numerous growth factors and cytokines, each of

which contributes to coupling bone dissolution (i.e.resorption) to new bone formation, orchestrate boneremodeling within a BMU (Figs 28.2 and 28.3). Pre-osteoblasts (pre-OBs), derived from mesenchymalstromal cells, and under the influence of a key tran-scription factor (Cbfa1, i.e. core binding factor I orRUNX2) represent target cells for initiation of theremodeling cycle (Martin & Ng 1994; Thissen et al.1994). Systemic and local factors, as well as signalsfrom osteocytes, enhance pre-OB differentiation,and this, in turn, leads to the synthesis and release of macrophage colony-stimulating factor (M-CSF)and receptor activator of nuclear factor kappa B ligand (RANKL) (Musey et al. 1993). These two

RANK

RANKL

M-CSF

IL-1, IL-6, IL-11

TNF, TGF-β

TGF-β

LTBP

Formation - 100 days

120 days

Resorption - 20 days

OPG

Activation

GH, IGF-I

IL-1, PTH, IL-6,

– E2

IGF-I, II

OsteocalcinOsteocalcin

BSAP

PICPMatrix

Proteases

Collagen

H+

Hydroxyproline

Calcium

IGF-I

IGF-II

IGFBPs

D-Pyr

CTXNTX

OC OB

Fig. 28.2 The bone remodeling cycle which is controlled by circulating and local growth factors and cytokines. Bonemarkers, including NTx, Ctx and D-Pyr, are fragments of collagen released during resorption. Formation markersincluding osteocalcin, BSAP and PICP are also noted by the osteoblast. BSAP, bone specific alkaline phosphatase; GH,growth hormone; IGF, insulin-like growth factor; IGFBPs, insulin-like growth factor binding proteins; IL, interleukin; M-CSF, macrophage colony-stimulating factor; OB, osteoblast; OC, osteoclast; PICP, procollagen peptide; RANK, receptoractivator of nuclear factor kappa B; RANKL, RANK ligand; TGF, transforming growth factor; TNF, tumor necrosis factor.

Page 427: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 411

peptides are necessary and sufficient for the recruit-ment of bone resorbing cells, i.e. the OCs. Once boneresorption occurs, calcium, collagen fragments andgrowth factors such as the insulin-like growth factors (IGFs) and transforming growth factors(TGFs), are released from the bony matrix. The latterfactors enhance the recruitment of OBs to the bonesurface, thereby setting the stage for collagen syn-thesis and matrix deposition/mineralization (Rosen& Donahue 1998). The entire remodeling cycle inhumans takes approximately 90 days, with themajority of time consumed by the elaborate processof bone formation and subsequent mineralization(Fig. 28.2) (Rosen & Donahue 1998). And, at eachstep, systemic hormones such as parathyroid hor-mone (PTH), estrogen, thyroxine and GH, influencethe timing and direction of remodeling in a three-dimensional space.

One of the most critical local and systemic growth factor influencing bone remodeling is IGF-I(Fig. 28.3). IGF-I and IGF-II, the IGFs, are majorcomponents of both the organic skeletal matrix andthe circulation. Indeed, the serum of most mammalscontains significant concentrations of both IGF-I

and IGF-II, bound to high and low molecular weightinsulin-like growth factor binding proteins (IGFBPs)(Ketelslegers et al. 1995). Similarly, the skeletalmatrix also is highly enriched with these growthfactors and other non-collagenous proteins, includ-ing all six IGFBPs and several IGFBP proteases. Inaddition, the type I IGF receptor is present on bothOBs and OCs.

It is now reasonably certain that skeletal IGFsoriginate from two sources: (i) de novo synthesis by bone forming cells (i.e. pre-OBs and fully differ-entiated OBs); and (ii) the circulation. In fact, someskeletal IGFs probably make their way into thematrix by way of specialized canaliculi and sinu-soids within the bone microcirculation (Rosen &Kessenich 1996; Rosen & Donahue 1998). IGFs,bound to IGFBPs, can also be found within the marrow milieu in close contact with the endostealsurface of bone. But, by most accounts, the vastmajority of IGF-I in bone is derived from localosteoblastic synthesis. Yet during active bone resorp-tion, as the matrix is dissolved, significant amountsof IGF-I and IGF-II are released from storage (i.e.binding to IGFBP-5 and hydroxyapatite) (Fig. 28.2).Subsequently, both IGFs recruit precursor OBs, and possibly early OCs, to the bone surface whereremodeling is occurring (Rosen & Kessenich 1996;Rosen & Donahue 1998; Heaney et al. 1999).

Circulating and skeletal IGF-I are profoundlyinfluenced by nutritional determinants and physicalactivity. Growth retardation, a major feature of pro-tein calorie malnutrition in children, is associatedwith significant declines in circulating IGF-I despiteenhanced GH secretion. Similarly elderly indivi-duals with poor protein intake have low serum IGF-I levels (Schurch et al. 1998). This almost certainly isdue to reduced mRNA half-life of IGF-I in the liver.But, regardless of the mechanism, IGF-I is located inthe final common homeostatic pathway affected bychanges in nutrient intake and energy balance. Assuch, this peptide emerges as an important medi-ator of the skeletal response to stress. For example, arecent study of elderly women who suffered a hipfracture (i.e. the end-stage of osteoporosis) supportthis contention. After a hip fracture there is a pro-found decline in serum IGF-I levels, probably asresult of poor nutrition and significant physicalinactivity, as well as a catabolic state (Schurch et al.

Fig. 28.3 The basic multicellular unit (BMU) consists of osteocytes connected by canaliculi to lining cells(cuboidal) and osteoblasts (ovoid). Osteoclasts aremultinucleated giant cells (see at the base of the lacuna)responsible for resorption of bone. The cellular responseto loading begins with signals from the osteocytes tolining cells and osteoblasts, resulting in activation of theremodeling sequence.

Page 428: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

412 chapter 28

1998). Levels of IGF-I can be partially restoredthrough the use of recombinant IGF-I administeredwith IGFBP-3 (Boonen et al. 2002). This treatmentregimen in elderly patients after hip fractures,resulted in less bone loss and significant improve-ment in functional outcomes (Boonen et al. 2002).This line of evidence supports the importance of a circulating mediator that affects skeletal respons-iveness to injury, particularly in relation to energystatus.

In children and young adults, exercise stimulatesGH secretion, which can result in higher levels ofserum IGF-I. This effect is blunted in adults suchthat serum IGF-I concentrations are not statisticallydifferent with sustained exercise regimens. How-ever, in respect to physical activity, anything thatreduces dietary ingestion of essential nutrients (i.e.prolonged physical activity with reduced intake, orcompulsive undereating) in adults or children willoverride GH stimulation of IGF-I in the liver andsignificantly reduce circulating IGF-I concentrations.

Exercise can also impact skeletal IGF-I expres-sion. Several studies have shown that fluid flow inosteocytes and OBs can increase the expression ofIGF-I mRNA (Srinivasan & Gross 2000). Repetitivegeneral physical activity not only increases IGF-Iexpression in muscle but also in the periosteum andprobably on the endosteal surface of bone. Thesechanges can have a profound effect on bone forma-tion tipping the remodeling balance in a favorabledirection, particularly during peak bone acquisi-tion. On the other hand, unloading removes thestimulus to bone formation, in part by making bonecells resistant to the actions of IGF-I (Sakata et al.2003) (see Hormonal and skeletal responses to long-term exercise section below).

The remodeling cycle is sensitive to changes inother nutrients which can profoundly affect growthfactors and cytokines in OBs. Phosphate balance isimportant for mineralization and low phosphatelevels trigger activation of 1α-hydroxylase keyingthe conversion of 25-hydroxyvitamin D to the activecompound 1,25-dihydroxyvitamin D. Conversely,high levels of phosphorus stimulate PTH secretionresulting in marked activation of remodeling andenhanced bone resorption. Low calcium intake, coupled with vitamin D deficiency, inhibits IGF-I

expression in bone, can trigger PTH release andalmost certainly is a principal factor in the secondaryhyperparathyroidism seen in elderly individuals. In addition, poor calcium diets and low vitamin Dlikely contribute to dampening of the skeletalresponse to loading (see Hormonal and skeletalresponses to long-term exercise section below). Vit-amin K is an essential co-factor for γ-carboxylationof osteocalcin, the most common non-collagenousprotein in bone. Osteocalcin is produced by boneforming cells that are highly differentiated, and thisprotein may be important in mineralization. Theexpression and release of osteocalcin has also beennoted in loading studies. Other trace elements suchas boron and strontium may affect bone cell func-tion in vitro, although their role in the remodelingcycle is uncertain. Similarly, low levels of magne-sium may influence bone cell activity in vitro, but itsrole in remodeling and the response to exercise isstill debated.

Aberrant remodeling

Aging and menopause are most frequently associ-ated with modest uncoupling of the BMU resultingin accelerated bone resorption compared to boneformation. Hence, over a lifespan, women can loseapproximately 42% of their spinal and 58% of theirfemoral bone mass (Rosen 2003). Surprisingly, ratesof bone loss in the 8th and 9th decades of life may becomparable or exceed those found in the immediateperi- and post-menopausal period of some women(Lacey et al. 1998; Robey & Bianco 1999). This is due to uncoupling in the bone remodeling cycle ofolder individuals resulting in a marked increase inbone resorption but no change or a decrease in bone formation (Martin & Ng 1994; Rosen & Donahue1998). However, the mechanisms that lead to anuncoupled bone remodeling unit, especially in theelderly, remain to be elucidated. Almost certainly,major changes in several hormonal factors (estro-gen, testosterone, GH) as well as nutrient intake,dramatically affect the skeleton either singly or in combination. The role of physical immobility on age-related changes in the skeleton remainsuncertain, but inactivity is likely to accelerate boneresorption in the elderly.

Page 429: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 413

Recent technological advances have made it easierto monitor the remodeling process by definingchanges in bone specific turnover markers in patho-logic states such as osteoporosis. Alterations in boneturnover can be detected by several biochemicalmarkers including bone resorption indices (e.g. urinary and serum N-telopeptide, C-telopeptide,and urinary free and total deoxypyridinoline) andbone formation markers (e.g. osteocalcin, procolla-gen peptide, bone specific alkaline phosphatase)(see Fig. 28.2). In general, bone turnover markers are significantly higher in older than younger post-menopausal women, and these indices are inverselyrelated to BMD (Beamer et al. 2000). For example inthe EPIDOS trial of elderly European females, the highest levels of osteocalcin, N-telopeptide, C-telopeptide and bone specific alkaline phosphatasewere noted for those in the lowest tertile of femoralbone density (Thissen et al. 1994). Also, increasedbone resorption indices were associated with agreater fracture risk independent of BMD (Thissenet al. 1994). For those women in EPIDOS with lowbone density and a high bone resorption rate, therewas a nearly fivefold greater risk of a hip fracture. Inrespect to exercise-induced changes in the skeleton,bone turnover markers may provide some insightinto the role of systemic factors in modulating the adaptive response to loading, particularly inrelation to bone resorption.

In contrast to a consistent pattern of high boneresorption indices with aging, bone formationmarkers in osteoporotic patients are more variable.Serum osteocalcin levels are high in some individu-als but this may be indicative of an increase in boneturnover rather than reflecting a true rise in boneformation (Thissen et al. 1994). On the other hand,bone specific alkaline phosphatase, and procollagenpeptide levels have been reported to be high, normal or low in elderly men and women (Musey et al. 1993). Bone histomorphometric indices in somepatients are also quite variable. Thus, althoughthere is strong evidence for an age-associated rise inbone resorption, changes in bone formation areinconsistent. Still, with aging and menopause, themajor skeletal abnormality is an uncoupling of the remodeling unit that leads to bone loss, alteredskeletal architecture and an increased propensity

to fractures. These determinants are particularlyimportant when considering the effects of bothacute and chronic exercise programs on the agingskeleton.

Weightlessness due to space flight has beenreported to induce the most rapid and highest rateof bone loss of any pathologic condition (Neuman1970). This process is a function of accelerated boneresorption and simultaneous suppression of boneformation which begins immediately after reachingzero gravity. Although hormonal replacement canameliorate part of this loss, restitution of normalbone mass only occurs with a normal gravity state.The role of physical inactivity that is not due toweightlessness or chronic bed rest, in the progres-sion of osteoporosis is less well defined. Severalstudies have shown that bed rest in healthy indi-viduals can be associated with uncoupling of forma-tion from resorption, with a high resorptive stateand suppressed formation (Chappard et al. 1995). Asimilar process likely occurs after a hip fracturewhen immobility is significant and bone loss fromthe contralateral hip can be significant even over arelatively short-time period (Sato et al. 2001).

Less attention has been paid to the role that theperiosteal surface may play in aberrant remodelingand the effects of inactivity on this region vis à vishormonal and local factors. A recent 20-year pro-spective study of post-menopausal Swedish womennoted that bone loss from the radius averagednearly 2% per year. However in these same women,periosteal circumference increased (Fig. 28.4) sig-nificantly, such that the cross-sectional moment ofinertia, an index of bone strength, actually increased(Ahlborg et al. 2003). This suggests that during theprocess of endosteal bone loss as a result of aging or hormonal deficiency, the periosteum attempts to compensate in order to maintain its inherentstrength (Duan et al. 2001). Currently it is not knownhow the periosteal envelope expands in response toloss of the inner aspect of bone, nor how it is sig-naled (Fig. 28.4) (Beck et al. 2000; Nelson et al. 2000).However, it should be noted that the two mostimportant regulators of periosteal growth are mus-cle activity and systemic IGF-I. Since the former canalso induce skeletal IGF-I expression in the perios-teum and skeletal muscle, this peptide may be very

Page 430: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

414 chapter 28

critical for the compensatory skeletal response withaging (Adams & Haddad 1996). Indeed, evidencefrom transgenic and knockout mice tell us that cir-culating IGF-I is important for modeling the skel-eton and providing it with a stimulus for optimalgrowth, particularly in the medial lateral dimension(Bikle et al. 2002). Since the periosteum is highly vascular and pericytes may be the origin of OBs inthis compartment, it is not too radical to considerthe GH–IGF-I hormonal axis, which may be dir-ectly or indirectly modulated by physical activity, as

the critical mediator of this compartment. Furtherstudies of this hypothesis are currently underway in several laboratories. Such studies will provide astrong rationale for studying the interface betweenphysical activity, systemic hormonal modulatorsand bone remodeling.

Hormonal and skeletal responses toacute exercise

The skeleton is an extremely dynamic organ thatresponds not only to systemic hormonal factors but to locally generated growth factors produced inresponse to applied forces (Table 28.1). As previ-ously noted, modeling, i.e. the process of growthand shaping, and remodeling, i.e. the process ofbone renewal, are responsive to both hormonalmediators and local stresses. More is known aboutthe skeletal response to systemic mediators than it is to loading. However, there are some basic bio-mechanical properties of the human skeleton thatapply to any form of loading and are synergisticwith hormonal factors that act on the BMU.

Bone exhibits a remarkable capacity to adapt tochanges in bone loading in order to optimize strengthwithout unduly increasing weight. Surprisingly, the skeleton does this by altering its mass, its exter-nal geometry and its internal micro-architecture, a

X-sectionalarea

BMD

Moment ofinertia

1.77

1.0

0.25

1.77

1.0

0.64

1.77

0.80

1.13

Fig. 28.4 The skeletal effects of aging, including the loss of endosteal bone. In response to declining trabecularbone, the periosteum increases in size. This enhances thebiomechanical properties of the bone and prevents totalfailure of the skeleton. BMD, bone mineral density.

Table 28.1 Systemic and local modulators of bone turnover: response to loading.

Hormone/ Systemic/ Local/ Loadinggrowth factor CNS skeletal effects Reference(s)

PTH +++ – Synergistic Rosen 2003; Chow et al. 1998, Neer et al. 2001GH +++ – Synergistic Forwood et al. 2001IGF-I +++ ++++ +++++ Rosen & Donahue 1998, Rosen & Kessenich 1996,

Bikle et al. 2002, Lean et al. 1996PGE2 (PGI) +++ ++++ +++++ Klein-Nulend et al. 1997Leptin +++ ++ ?? Takeda et al. 2002Neuropeptides +++ ++? + Elefteriou et al. 2003, Takeda et al. 2002, Mason et al. 1997TGF-β + +++++ + Srinivasan & Gross 2000Estradiol ++++ ++ +/Synergistic Williams et al. 1995, Dueck et al. 1996, Otis et al. 1997,

DiPietro & Stachenfeld 1997, Klibanski et al. 1995, Grinspoon et al. 2003, Smith & Rutherford 1993

Testosterone ++++ + Synergistic Rosen 2003Cortisol ++++ ? Antagonistic Chrousos & Gold 1992

CNS, central nervous system; GH, growth hormone; IGF-I, insulin-like growth factor I; PGE2, prostaglandin E2; PGI, prostacyclin; PTH, parathyroid hormone; TGF-β, transforming growth factor-β.

Page 431: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 415

concept that has been around for more that 100years (Boyden et al. 2002). What has emerged morerecently, however, is a better understanding of thefactors that influence the skeletal response to load-ing, including sensitizing hormones such as PTHand GH (Turner et al. 1997). To begin with, loadsapplied to the skeleton are called stresses (i.e. forceper unit area), while strain represents the measureof skeletal deformation in response to a stress (i.e.change in length divided by its original length). It isthe strain per se that generates an adaptive responseof bone to loading. Adaptation of hard tissues tostress is achieved by changes in bone resorption andbone formation. Bone mass, geometry and trabecu-lar orientation are altered as an adaptive responseand these, in turn, are modulated by systemic factors.

Not all of the adaptive responses to stress areequal because of the geometric properties of bone,but there is a direct relationship between the magni-tude of strain, and the appropriate skeletal response(Rubin & Lanyon 1985). The cellular mechanismresponsible for that relationship is not known butinvestigators have posited that there must be a‘mechnostat’ which regulates both periosteal forma-tion and endosteal resorption. If bone is loaded with more than 2500 microstrain, modeling occursat both sites, thereby allowing the bone to be moreresistant to deformation (Rubin & Lanyon 1985). In contrast, when strain is reduced, modeling isinhibited and endosteal remodeling with resorptionoccurs. The ‘putative’ mechanostat has never beenidentified, but theories abound about as to its loca-tion and its mechanism of action. Most agree thatosteocytes, buried within the cortex of bone but connected to resting surface cells, sense fluid shiftsand gravitary forces (Noble & Reeve 2000). These‘strains’ trigger release of cellular factors whichtravel by way of canaliculi to the surface of bone to activate resting OBs or pre-OBs (see Fig. 28.3)(Turner 1999). This communication allows marrowstromal cells to be recruited and begin the differ-entiative process necessary for bone formation.Recently, evidence has emerged from genetic studiesof a family with high bone mass but normal boneshape, that a previously unidentified pathway inOBs may have ‘mechanostat’ like properties. Lipo-protein receptor protein 5 (LRP-5) is a ubiquitous

membrane receptor which is coupled to the frizzledreceptor on OBs and is activated by a family ofgrowth related peptides called Wnts. These growthfactors can stimulate bone formation and mineral-ization as well as cell proliferation, and are nowthought to work through a canonical pathway incells (Gong et al. 2001). LRP-5 interaction with theWnt frizzled ligand receptor complex results ininhibition of β-catenin phosphorylation by glycogensynthetase kinase 3 (GSK-3) (Kato et al. 2002). GSK-3facilitates the ubiquitin mediated breakdown of β-catenin, while LRP-5 acts to prevent this break-down allowing for translocation of β-catenin intothe nucleus where it interacts with a family of tran-scription factors. An activating mutation of LRP-5 is responsible for the ‘high bone mass’ phenotype,which has been reported in several families withhealthy individuals but with BMD values 3–5 stand-ard deviations above normal, yet normal boneremodeling (Boyden et al. 2002; Little et al. 2002).Very recently investigators have noted that mutantLRP-5 expression was up-regulated by mechanicalstimulation, placing this system in the forefront ofcandidates for the elusive mechanostat (Bex et al.2003).

The type of load applied defines the particularadaptive response of the skeleton in co-ordinationwith hormonal mediators. Three critical factorsrelated to physical activity modulate strain in thehuman skeleton: frequency, duration and intensity(Rubin & Lanyon 1984). The latter is probably themost important since intensity generates load. Forexample, gymnasts can experience ground forcestwelve times their body weight, while runners expe-rience only about three to five times their weight(Grimston 1993; Grimston et al. 1993; Bassey et al.1998). As such, the greatest increases in BMD amongactive exercisers occurs at the point of impact (i.e.the hip) and is most pronounced in gymnasts ratherthan runners or walkers.

The frequency and duration of physical activityare also important. Individuals who perform jumpsfrom short heights rapidly develop a peak force onimpact; these subjects have greater increases in bonedensity than do runners, and bone formation para-meters have been directly linked to strain rate, inde-pendent of force (Morris et al. 1997; Fuchs et al. 2001).

Page 432: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

416 chapter 28

In addition to the rate of change in force, evidencehas emerged from both animal and human studiesthat the frequency in which sound waves are gener-ated by a floor-based machine can affect the rate ofbone formation and the acquisition of bone mass(Qin et al. 2003). Whether this represents a purelyskeletal effect, or one which is co-ordinated throughskeletal muscle remains to be determined. Howeverit does provide new insights into the bone modelingprocess. It is unknown whether there are additionalhormonal factors which contribute to or are synger-stic with this effect.

Certain hormones can facilitate the ability ofmechanical loading to increase bone formation. Thetwo major ones are PTH and GH. Removal of theparathyroid glands in rats abolishes the skeletal sensitivity to mechanical loading. Daily replace-ment with PTH restores that mechanical sensitivityalthough the mechanism responsible for this effectis unknown (Chow et al. 1998). One candidate as alocal mediator is IGF-I, which is induced by PTHdirectly through increases in skeletal gene expres-sion. IGF-I null mice do not exhibit any anabolicresponse to PTH, nor do they show changes inmarkers of bone formation (Bikle et al. 2002). Theother important hormonal mediator is GH, anotherpowerful inducer of local IGF-I. Lewis dwarf ratsthat do not have an intact GH–IGF-I axis do notrespond to skeletal loading, but this mechano-sensitivity can be restored with GH replacement(Forwood et al. 2001). Once again, it is not clear howGH mediates this effect, although replacementinduces a significant rise in both local and systemicIGF-I.

The importance of a potent ‘modeling’ factor suchas GH cannot be understated in respect to the grow-ing skeleton of pubertal children. Exercise-inducedchanges in bone mass are most pronounced duringthe late prepubertal and pubertal growth phases(Bass 2000). Indeed, post-puberty, exercise-inducedchanges in bone mass are relatively minor, althoughthe effects on the periosteum require further stud-ies in older individuals. Recently, PTH has beenapproved for the treatment of post-menopausalosteoporosis based on a large randomized placebocontrolled trial (RPCT) of post-menopausal women(Neer et al. 2001). It increases bone mass by enhanc-

ing periosteal expansion and stimulating endostealbone formation. Its potential synergy with physicalactivity has not been tested in vivo, but studies arecurrently being planned.

The acute response to physical activity or loadingis systemic and local. The hormonal modulatorsinclude changes in classical stress hormones such as cortisol and epinephrine. There are also bi-directional responses in the immune system relatedto neuroendocrine function. GH levels rise, particu-larly in younger individuals, although changes inIGF-I concentrations are not noticeable and are bal-anced by energy needs and nutrient intake. Insulinlevels are generally suppressed while glucagon isincreased with physical activity. Accompanying therise in heart rate is the release of central nervous system trophic factors such as neuropeptide Y, aswell as systemic and local cytokines tied to theimmune response, including interleukin-1 (IL-1),IL-4, IL-6, interferon-γ and IL-11 (Chrousos & Gold1992). There are also significant changes in circulat-ing white blood cells as a response to these variousimmune modulators and sympathetic activity isalso increased, leading to changes in blood pressureas well as heart rate.

All of the above modulators have also beenshown to impact the skeleton in one fashion oranother when exposure is chronic. The importantrole of neuropeptides in controlling OB function hasrecently emerged, as has the possibility that leptin,an adipocyte-derived factor, plays a role in sup-pressing bone formation and mediating hypothala-mic events related to obesity and starvation (Blum et al. 2003; Cock & Auwerx 2003; Elefteriou et al.2003). Sympathetic overactivity is responsible forthe syndrome of reflex sympathetic dystrophy, andβ-adrenergic blockers can prevent bone loss inrodents following ovariectomy (Takeda et al. 2002).However, none of these mediators play a major rolein the acute response of the skeleton to loading. Thatadaptation is part of the mechanostat and is local-ized to the osteocyte and OB. Moreover, changesthat occur in the skeleton as a direct response toloading happen over relatively short-time intervals(e.g. minutes), well before hormonal modulators are active. On the other hand, sustained changes insystemic factors are probably responsible for the

Page 433: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 417

more balanced skeletal response to physical activityseen with long-term physical activity.

Osteocytes, as noted previously, are buried deepwithin the cortex of the skeleton (see Fig. 28.3). Theyrepresent old OBs that have been entombed withinthe matrix these cells have generated (Noble & Reeve2000). Despite their relatively modest metabolicrate, these terminally differentiated cells can sensefluid sheer stress as mechanical loading. These cellsalso communicate with bone lining cells and OBsthrough gap junctions by way of canaliculi whichcan carry specific growth factors and cytokines (seeFig. 28.3). As a result, loading can transduce a relat-ively rapid message to the master control cells of theBMU in order to initiate remodeling. There is someevidence to suggest that lining cells can even makematrix in response to mechanical loading by de-differentiating into OBs.

The earliest changes in the osteocyte after load-ing, is an increase in ocsteocytic glucose-6 phos-phate dehydrogenase activity (Lean et al. 1996). Thisoccurs within minutes of the stimulus. At the same time, OBs demonstrate an increase in intracel-lular calcium, probably as a result of activation ofthe phosphatidylinositol 3-kinase (PI3K) pathwaywhich mediates intracellular calcium release. Thesechanges result in stimulation of the mitogen activ-ated protein (MAP) kinase signaling pathway, a keycircuit for activating gene transcription. At 30–60 min after induction of stress on the bone, liningcells and OBs begin to express c-fos, an importantproto-oncogene necessary for both OB and OC function. IGF-I expression is up regulated in the OB at 1 h (Lean et al. 1996). Other factors clearly contribute to the increase in OB activity, includingprostaglandins, TGF-β, neuropeptides and severalmatrix proteins that can activate integrins andenhance cell movement, particularly in marrowstromal cells.

Prostaglandins likely play an important role inthe activation of bone remodeling with loading.Inhibition of prostaglandin synthesis by non-steroidalanti-inflammatory drugs suppresses bone forma-tion in vivo. The two most active prostaglandinsreleased during this process are prostaglandin E2(PGE2) and prostacyclin (PGI2) (Klein-Nulend et al.1997). These compounds are synthesized both in the

osteocyte and OB and may be major signals forrecruitment of OB precursors from marrow stemcells. Selective inhibition of the inducible prostag-landin synthase (COX-2) results in a greater sup-pression of loading induced bone formation than anon-selective blockade, although the clinical signific-ance of this is currently unknown (Forwood 1996).

As noted earlier, neuropeptides are importantsystemic mediators of the stress response. For example, neuropeptide Y is downstream of leptin in the hypothalamus and can cause bone loss whenadministered into the cerebral ventricles of mice.Also neuropeptide Y deficient mice have a markedincrease in trabecular bone as do conditional knock-out mice that have deletions of neuropeptide Y limited to the hypothalamus. In addition to the sys-temic and central nervous system effect, recent evid-ence suggest these proteins may also be importantin the acute local response to loading. A glutamatetransported has been identified in bone tissue and is down regulated immediately after mechanicalloading (Laketic-Ljubojevic et al. 1999). Serotoninreceptors are present on osteocytes and OBs, particu-larly in the periosteum suggesting that mechano-transduction may in part be mediated through thispathway (Mason et al. 1997).

Two other major processes occur with loading at the cellular level: prevention of apoptosis of OBsand osteocytes as a result of local growth factors(IGF-I, leptin, IL-6, TGF-β) and the release and elab-oration of nitric oxide, an important mediator of OB activity and an inhibitor of osteoclastic function.Nitric oxide may be involved in mechanotransduc-tion although the mechanism has not been defined(Turner et al. 1996). It has recently been shown todown-regulate RANKL expression in stromal cells,a key factor in osteoclastogenesis (Rubin et al. 2003).Nitric oxide also can bind to guanylyl cyclase stimu-lating cyclic guanosine monophosphate (cGMP),another intracellular mediator that can regulategene transcription.

In sum, a host of local growth factors relativelyrapidly enhance the number of OB progenitors andterminally differentiated cells at the remodeling sur-face in response to loading. Interestingly, hormonalmediators of the acute stress response that are activeduring the first minutes of physical activity do not

Page 434: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

418 chapter 28

modulate the adaptive response in the skeleton toloading. But, ironically, their presence and regula-tion in the local bone milieu plays a major role in thechronic adaptive response to loading. Cytokines,growth factors, neuropeptides, prostaglandins andnitric oxide contribute to an increase in bone forma-tion with physical activity.

Hormonal and skeletal responses tolong-term exercise

As noted earlier, the skeleton is ideally suited torespond to mechanical loading by signal trans-duction resulting in modeling of the periosteum andtrabecular reorientation. Hormonal changes as aresult of physical activity play a minor role at the tissue level in accommodating the acute adaptiveresponse. However, hormonal cues are particularlycritical for regulating the process of bone remodel-ing, an essential element of bone preservation inadult life. As such, the skeletal adaptation to mech-anical loading which results from repetitive muscleactivity is balanced by the long-term changes inendocrine modulators such as the gonadal steroids,cortisol and neuropeptides. These changes will bediscussed in this section.

Most investigators believe, although there are no long-term prospective data, that a lifetime ofphysical activity benefits the skeleton (Bouxsein &Marcus 1994). This, indeed is the official position ofthe American College of Sports Medicine (AmericanCollege of Sports 1998). However, skeletal adapta-tion to loading is very site specific, so that generalit-ies about the long-term benefits of physical activityon bone must be avoided. Moreover, as noted earlier, the beneficial effects of loading the skeletonthrough exercise are much more pronounced beforeand during puberty than later in life (Bass 2000).Hence, changes in BMD, as measured by dualenergy X-ray absorptiometry (DXA), are likely to bemuch greater in children than adults. In fact, most ofthe trials related to physical activity in adults havedemonstrated only very modest changes in BMD ofthe spine or hip, despite, in some cases, significantmusculoskeletal loading. This difference almostundoubtedly relates to the fact that loading of theadult skeleton preserves bone mass by slowing rates

of resorption and increasing slightly bone forma-tion, a result of changes on the endosteal surface of bone. On the other hand, changes in skeletal mass with loading in children can have a profoundeffect on the growing skeleton, particularly on theperiosteal surface. For example, tennis and squashplayers who begin playing in their preadolescentyears have a two to fourfold times greater differ-ence in radial BMD between their playing arm andnon-playing arm compared to those who start afterpuberty (Kannus et al. 1995). Similarly, young pre-pubertal girls who undertook a regimen of regularweight-bearing activity plus jumping for 10 monthshad a nearly 6% increase in femoral bone mineralcontent compared to an 8-month intervention inadolescents (age 14.2 years) of a similar nature(Witzke & Snow 2000; Fuchs et al. 2001). In that lat-ter study, there was virtually no change in bone mineral content or density at any site except thefemoral trochanter. Thus site specificity and age are major determinants of the chronic response toloading. Finally, it should be noted that a major factor that is difficult to quantify, particularly inlarge-scale trials, is the amount of background phys-ical activity occurring before or during the conductof the study. Although questionnaires are useful inassessing the amount and frequency of backgroundactivity, these are certainly imprecise and subject to considerable variability, particularly in childrenwhere normal daily activities are quite pronounced.

However, a word of caution is necessary in rela-tion to the assumption that loading the skeleton hasa more pronounced effect in growing childrenrather than adults. Virtually all of the longitudinalstudies showing changes in bone mineral content or BMD in adolescence or preadolescent childrenutilized areal measurements derived from DXA.This technique is two-dimensional and very sizedependent; thus areal BMD would be expected tomarkedly increase during the peak skeletal growth.This complicates interpretation of the longitudinaldata and bespeaks the need for studies looking atvolumetric or three-dimensional BMD to determinewhether exercise-induced benefits to the skeletonare purely growth mediated or in fact are relateddirectly to greater mineralization and matrix deposi-tion, the critical determinants of BMD.

Page 435: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 419

The chronic hormonal and skeletal response tosustained physical exercise is complex and variable.First it should be noted that there are numerous animal studies demonstrating a sustained skeletalresponse to chronic loading. Lanyon, Rubin andothers have convincingly demonstrated in avianulna that consecutive cycles of loading resulted in astrain magnitude which enhanced endosteal boneformation (Rubin & Lanyon 1985). In fact, 36 cyclesper day (2050 microstrain at 0.5 Hz) increases boneformation and further increases in cycle number donot increase bone mass (Rubin & Lanyon 1984).However, 1000 microstrain at 1 Hz led to a dose-dependent increase in periosteal bone deposition(Rubin & Lanyon 1985). Yet, exercise-induced stimu-lation of bone cells in the rib following weight-bearing exercise does not induce bone formation.Furthermore, unloading of animal bones withmicrogravity, hindlimb immobilization, spinal cordinjury, or sciatic neurectomy, result in significantbone loss. These studies confirm significant com-partmental differences (trabecular versus periosteal)as well as site specificity in regards to chronic load-ing or unloading of the skeleton.

Human adult studies have confirmed the resultsin animals, although the size effect related to exercise intervention is much lower. For exampleexercise training in young premenopausal womenenhances areal BMD in a site-specific manner.Resistance training and weight-bearing exercisesresult in a very modest increase in lumbar spine,femoral and calcaneal BMD (Bassey et al. 1998).And, there does not appear to be a significant dosedependency in women for these changes, althoughthose studies are not as neatly designed nor as con-clusive as animal studies. Older premenopausalwomen also show stabilization of areal BMD over a12-month period in response to chronic loading;however, these changes are not associated with astatistically significant increase in bone mass, sug-gesting that the effect size of the intervention maydecrease with increasing age (Pruitt et al. 1992;Bassey et al. 1998).

The response of bone to chronic exercise in post-menopausal women is somewhat conflicted. Inearly post-menopausal women without estrogenreplacement, resistance exercise either increased or

maintained BMD in the spine and sometimes in the femur, but failed to prevent bone loss at othersites (Kohrt et al. 1997). On the other hand, womenreceiving hormone replacement therapy (HRT) whounderwent a series of chronic exercise programsgenerally showed a marked increase in spine, totalbody and femoral BMD (Kohrt et al. 1997, 1998). The combination of exercise and calcium supple-mentation in post-menopausal women may begreater than exercise or calcium supplementationalone, as reported in a meta-analysis performed bySpecker (1996). As such there appears to be strongevidence that the optimal skeletal response to exer-cise in post-menopausal women requires adequatecalcium supplementation and HRT (Kanders et al.1988).

Exercise intervention studies in men are some-what more conflicted than in women. Training ofrecreational male athletes between the ages of 25–52years for 3 months with walking or running failed todemonstrate an increase in bone mass at the spine,humerus, femur or calcaneus. Similar but smallerstudies in older men also failed to show any changeswith a 1-year-long exercise regimen. On the otherhand, in army recruits completing 14 months ofintensive physical training, leg bone mineral con-tent increased 12.4% (Jones et al. 1989). This differ-ence may be a function of intensity, age or site ofmaximal loading. However it does bespeak thetremendous heterogeneity between studies, and thedifficulty in performing systematic reviews usingmeta-analysis for exercise trials.

One of the major limitations of loading trials inhumans relates to the endpoint being studied; i.e.areal BMD. DXA measurements of the spine or hipmay not capture the true effect of skeletal loading on bone mass. As noted previously, the skeleton iscomposed of both cortical and trabecular compon-ents. Force induced changes in the periosteal andendosteal envelope that surround these compart-ments are not easily captured by an areal approx-imation of bone mass. Indeed, changes in shape as a result of skeletal modeling can not be assessed byconventional DXA. Recently, evidence has emergedthat changes in areal BMD with anti-osteoporosistherapies, reflect only to a small degree the degree ofrisk reduction for subsequent fracture (Cummings

Page 436: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

420 chapter 28

et al. 2002). In other words, the variance around frac-ture risk in relation to any intervention is muchgreater than that attributed to changes in BMD, andalmost certainly reflects qualitative changes in bothcompartments of the skeleton. Hence, exercise inter-vention studies that employ BMD as a final end-point may underestimate the true effect on loadingon bone strength and fracture risk. Since there arestill no exercise trials with fracture as an endpoint,the degree to which changes in bone density reflectultimate skeletal strength remains uncertain. Newerimaging technology such as micro computed tomo-graphy (µCT), may allow for in vivo scanning of trabecular bone, while magnetic resonance imaging(MRI) can precisely delineate periosteal circum-ference, cortical thickness, trabecular number andconnectivity, muscle mass and marrow fat. Theseendpoints may improve our capacity to define theeffects of loading on the axial and appendicularskeleton, particularly in respect to aspects of bonegeometry and quality.

Regardless of the imaging tool, BMD representsthe sum of many factors over the lifetime of an individual. Particularly relevant to defining overallchanges in bone quantity is the role of circulatinghormones, cytokines and growth factors. Indeed, inosteoporotic states, delineation of hormonal status(estrogen in women, testosterone in men) repres-ents the first step in establishing the etiology of lowbone mass. Moreover, other endogenous hormonessuch as PTH, 1,25-dihydroxyvitamin D, 25-OH vitamin D and thyroxine, all contribute to changesin bone remodeling and are important in deter-mining the length of the remodeling cycle and itssubsequent balance. Similarly, circulating cytokinessuch as IL-6 have been shown to be important in thepathogenesis of age-related bone loss and primaryhyerparathryoidism (Nakchbandi et al. 2002). Asdiscussed previously, IGF-I circulates in high con-centrations and plays a major role in skeletal model-ing and cortical bone mass (Rosen & Donahue 1998).Acromegaly, a condition of excess GH and IGF-I, isassociated with large bone volumes and greaterBMD (Rosen & Donahue 1998). By contrast, peoplewith conditions such as hypopituitarism have smallbones, increased skeletal fragility and reducedBMD. These pathologic states are useful in definingthe role of circulating factors on the skeleton and

provide some basis for our understanding of thebalance between local and systemic factors. Theeffects of a chronic exercise regimen on systemic factors, such as gonadal steroids, remains a criticalaspect of the ultimate skeletal response to loadingand must be considered within the context of theentire organism.

Much like the osteoporotic states of aging, thepathologic syndrome now referred to as the femaleathelete’s triad, has provided investigators withsignificant insight into the interface between circu-lating hormones, body mass and skeletal adaptationto exercise. This condition is defined by amenorrhea(or hormonal disturbances), anorexia and low BMD(Fig. 28.5) (Drinkwater et al. 1984; Marcus et al. 1985;Tomten et al. 1998). It is most commonly seen inyoung adult women. To appreciate the adverseeffects of this syndrome on the skeleton and its relationship to circulating hormones, an overviewof the changes that result from dietary restraint andexcessive physical activity is necessary.

The hypothalamic–pituitary axis is the principleresponder to stress related to dietary changesand/or physical activity. Two major hypothalamicfactors, corticotrophin-releasing hormone (CRH),and gonadotropin-releasing hormone (GnRH) aresignificantly affected by energy balance associatedwith activity patterns and nutrient availability.

Low E2Low LH/FSH

Decreased energy balanceLow T3Low IGF-ILow body fat/BMILow bone mass

Increased risk of fracture,Increased morbidity, mortality

Amenorrhea Anorexia

Fig. 28.5 The female athlete triad. This syndrome,common in young athletic women, represents acompelling interface between hormones, exercise andbone adaptation. BMI, body mass index; E2, estradiol;FSH, follicle-stimulating hormone; IGF-I, insulin-likegrowth factor I; LH, luteinizing hormone; T3,triiodothyronine.

Page 437: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 421

CRH controls the acute stress response; it in turnstimulates adrenocorticotropic hormone (ACTH)release from the pituitary gland, subsequently lead-ing to cortisol production in the adrenals (Webster et al. 2002). CRH is released into the median emin-ence of the hypothalamus through the portal circu-lation from neuronal cells of the parvaentricularnucleus. It, combined with vasopressin, stimulatesACTH. Subsequently the increase in cortisol pro-duction enhances available energy stores by drivingprotein catabolism, stimulating fatty acid releasefrom adipose tissue and enhancing gluconeogen-esis (Darmaun et al. 1988; Chrousos & Gold 1992).Cortisol also modulates the physiological effects ofvarious catecholamines and exerts negative feed-back control on the hypothalamic–pituitary axis. Allthese processes are integral for an individual requir-ing major energy sources for the brain. But, long-term stimulation of this system can result in excesscortisol release via the hypothalamic–pituitary axis,leading to increased serum cortisol concentrationsand deleterious effects on muscle and bone.

The effects of long-term stress or overexer-cise on the GnRH, luteinizing hormone/follicle-stimulating hormone (LH/FSH) system have alsobeen studied in some detail. GnRH pulsatility is keyto the appropriate stimulation of both LH and FSH,the central regulators of ovarian steroidogenesis. Acommon factor in female athletes with amenorrheaor oligomenorrhea is a disruption in LH pulsatility,almost certainly originating from the GnRH pulsegenerator (Loucks et al. 1992). The precise mechan-ism for this early loss of pulsatility however remainsspeculative. But a likely scenario is that a reductionin energy availability prevents the hypothalamicpulse generator from providing the appropriate signal for LH/FSH release. It seems likely that opti-mal menstrual function almost certainly requires aminimum energy requirement; when that thresholdis not attained, loss of gonadotropin pulsatilityresults in a downstream shutdown in gonadotropinrelease. Several lines of evidence support this thesis.Loucks & Callister (1993) demonstrated that, ineumenorrheic sedentary women, reduced energyintake over 5 days resulted in impaired LH pulsatil-ity and a fall in trioiodothyronine. This drop occurredat a threshold energy level of 20–25 kcal·kg–1 (84–105 kJ·kg−1) of lean body mass. Similarly, Williams

et al. (1995) showed that active eumenorrheicwomen experienced suppressed LH pulsatility afteronly 3 days of physical training when dietaryenergy intake was reduced. This was completelyreversed with appropriate dietary intake. Finally,Dueck et al. (1996) showed in three amenorrheic andthree eumenorrheic women that increasing energyintake by 350 kcal·day–1 (1463 kJ·day−1) and reduc-ing training by 1 day per week was associated with anet increase in energy availability of 250 kcal·day–1

(1045 kJ·day−1). At the end of 15 weeks, all six athletes were retested and the amenorrheic athleteshad resumption of LH pulsatility as well as a reduc-tion in cortisol secretion.

Chronic changes in the hypothalamic pituitarysystem centered on GnRH and CRH can have majoreffects on the skeleton and almost certainly are partof the pathogenetic mechanisms associated with the female athelete’s triad (DiPietro & Stachenfeld1997; Otis et al. 1997). Weight loss, intentional orunintentional due to an energy deficit, contributesto a reduction in BMD by altering the mechanicalproperties of bone in relation to other body compon-ents and lowering systemic or local estradiol con-centrations. For example, reduced total body weightmeans less gravitary forces placed on the skeleton,thereby resulting in less strain, and a reduced adapt-ive response. Moreover, fat is a major source ofperipheral estrogen production as a byproduct oftestosterone catabolism. Marrow and circulatingestradiol levels drop in response to a decrease in the total number of fat cells, or their cell density atperipheral sites such as the bone marrow. Also, areduction in body fat may reduce the number ofavailable marrow precursor cells that could even-tually become preosteoblastic bone forming cells.Regardless of the mechanism, a drop in estradiol inyoung women results in increased bone resorption,particularly on the endosteal border. If low estradiollevels persist, resorption exceeds formation andbone is lost. Moreover, the beneficial effects of phys-ical activity on the formation side may be dimin-ished or abolished by the lack of estrogen, as notedpreviously for post-menopausal women.

Surprisingly, not all of the deleterious effects on BMD in amenorrheic athletes can be attributed to low circulating or skeletal estradiol. In fact,restoration of BMD is unlikely to occur in anorexic

Page 438: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

422 chapter 28

women even with HRT unless body weight isrestored to at least 70% of ideal weight (Klibanski et al. 1995). Thus energy restriction, low body weightand low body fat combine to drastically alter notonly the set-point for the two major hypothalamicfactors noted earlier (GnRH and CRH) but also for other factors that are nutritionally or energydependent. These include triiodothyronine, dehy-droepiandrosterone (DHEA), GH, IGF-I, IGFBP-1and several cytokines. In fact, parenteral IGF-I ther-apy can at least partially reverse the skeletal mani-festations of anorexia nervosa, particularly whencombined with an anti-resorptive agent (Grinspoonet al. 2003).

In summary, the long-term effects of a vigorousexercise program on the skeleton are dependent on the energy threshold, nutrient intake, type ofphysical activity, as well as local and circulatoryhormones. In general, weight-bearing exercises promote bone formation and can, particularly inyounger individuals, enhance bone mass throughits adaptive response to strain. However, excessiveexercise programs that can lead to negative energybalance and weight loss result in a significantdecline in estradiol levels such that the skeleton is atsignificant risk for rapid bone loss. The end result ofthese processes include a higher risk of fracture andsignificant immobility and morbidity. Moreover, ifthe female athlete is young enough (i.e. 15–19 yearsof age), the loss of bone mass during this intervalmay not be restorable with return of menses andresumption of normal body weight. Since exer-cise has become a more frequent habit among allindividuals, and thinness is certainly in vogue, par-ticularly in female college-aged students, the com-bination of several lifestyle factors in young femalesmay have a devastating impact on their long-termskeletal health, despite the short-term benefitsrelated to skeletal adaptation.

It should be noted that there is likely to be animportant gender effect on bone related to long-term exercise such that women appear to be moresusceptible to the adverse effects of sustained negative energy balance than men. However, thehypothalamic–pituitary–adrenal (HPA)–gonadalaxis in long-term male athletes has not been studiedas extensively as the HPA axis of women athletes. Infact, the clinical counterpart of the female athlete’s

triad in men may not exist per se since there are noeasy markers for hypothalamic dysfunction short of reduced libido. On the other hand, at least twogroups have now demonstrated that male triath-letes who are in the age range of 20 –40 years havereduced total and free testosterone compared tonon-exercising control men (Wheeler et al. 1984;Smith & Rutherford 1993). Moreover, althoughexercise programs in elderly men have virtually noeffect on circulating testosterone, sex hormonebinding globulin levels are higher in exercisers thancontrols, thereby affecting free testosterone con-centrations (Cooper et al. 1998).

Clinical correlates related to theinteraction of exercise, hormones andbone remodeling

Exercise is considered by many to be essential for overall well-being. The general benefits clearlyoutweigh the risks, and therefore prescriptions for healthier bones should include a daily exerciseregimen. Unfortunately, the overall impact of anexercise program on fracture risk in older men orwomen is not known. It is safe to assume that exer-cise increases muscle performance and possiblymuscle mass. This alone may be enough to reducefalls in older individuals, and thereby indirectlyreduce fractures. Loading of the skeleton with theresultant adaptive response is optimized in situ-ations where there is adequate gonadal steroids,although there are likely to be skeletal benefits forolder individuals that are gonadally insufficient.

Risks of a sustained exercise program are relat-ively small and relate to problems loading areas ofthe skeleton that are not adaptable to stress, andcrossing a theoretic threshold where energy balancebecomes negative in the face of greater muscu-loskeletal activity. The former issue has becomemore important for the armed services, where theprevalence of stress fractures among new recruitsrange from 10–20% (Bennell et al. 1996a, 1996b). Inmany cases these cortical fractures cannot be relatedto areal BMD, but rather are a function of trainingand readiness. In addition, for young female armyrecruits, stress fractures of the pelvis and tibia aremuch more prevalent than in male recruits. Thismay be a function of loading in regions of the skel-

Page 439: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 423

eton, particularly the periosteum, that are not used tohandling repetitive forces. These fractures cost thearmed services dearly in respect to combat readi-ness, as well as the financial implications related to caring for these individuals. Factors that predictstress fractures in military recruits or elite athleteshave not been well delineated, but clearly are some-what independent of bone density or the strength of the force applied (Bennell et al. 1996a, 1996b).Qualitative measures of bone, as determined bynewer imaging technologies, may provide someclues for the future identification of those indivi-duals susceptible to injury. Similarly, because thereappears to be a heritable component to stress frac-tures, genetic studies may identify those high-riskindividuals prior to the onset of training.

Less clear cut are the clinical correlates related tothe deleterious effects of exercise on energy balance,particularly in the young female athlete. Oral con-traceptives work in some subjects who have the

female athlete’s triad; however, restoration ofweight remains the best prognostic factor for re-sumption of menses and improvement in skeletalhealth. Newer studies have been initiated withDHEA, a weak adrenal androgen, to determine ifthis compound is more palatable and as effective asestrogen in stopping bone resorption and enhancingbone formation (Gordon et al. 2002). Other trialswith newer anabolic agents are being considered.

As noted previously, aging is associated withsignificant bone loss, accelerated bone resorptionand a high risk of fractures. Defining the optimalthreshold for loading the skeleton and the resultantenergy balance will be mandatory in the future ifexercise is to be used as a prescription for betterbone health. In the meantime, a major push isneeded to fully understand the fine balance betweenlocal and systemic hormones and skeletal remodel-ing, particularly in respect to exercise programs andtheir more global application.

References

Adams, G.R. & Haddad, F. (1996) Therelationships among IGF-1, DNAcontent, and protein accumulationduring skeletal muscle hypertrophy.Journal of Applied Physiology 81(6),2509–2516.

Ahlborg, H.G., Johnell, O., Turner, C.H.,Rannevik, G. & Karlsson, M.K. (2003)Bone loss and bone size aftermenopause. New England Journal ofMedicine 349(4), 327–334.

American College of Sports MedicinePosition Stand (1998) Exercise andphysical activity for older adults.Medicine and Science in Sports and Exercise 30(6), 992–1008.

Bass, S.L. (2000) The prepubertal years, auniquely opportune stage of growthwhen the skeleton is most responsive toexercise? Sports Medicine (Auckland, NewZealand) 30(2), 73–78.

Bassey, E.J., Rothwell, M.C., Littlewood,J.J. & Pye, D.W. (1998) Pre- andpostmenopausal women have differentbone mineral density responses to thesame high-impact exercise. Journal ofBone and Mineral Research 13(12),1805–1813.

Beamer, W.G., Donahue, L.R. & Rosen, C.J.(2000) Insulin-like growth factor I andbone: from mouse to man. GrowthHormone & IGF Research 10 (suppl. B),S103–S105.

Beck, T.J., Looker, A.C., Ruff, C.B.,Sievanen, H. & Wahner, H.W. (2000)Structural trends in the aging femoralneck and proximal shaft: analysis of theThird National Health and NutritionExamination Survey dual-energy X-rayabsorptiometry data. Journal of Bone andMineral Research 15(12), 2297–2304.

Bennell, K.L., Malcolm, S.A., Thomas, S.A.et al. (1996a) Risk factors for stressfractures in track and field athletes. A 12-month prospective study. AmericanJournal of Sports Medicine 24(6), 810–818.

Bennell, K.L., Malcolm, S.A., Thomas, S.A.,Wark, J.D. & Brukner, P.D. (1996b) Theincidence and distribution of stressfractures in competitive track and fieldathletes. A 12-month prospective study.American Journal of Sports Medicine 24(2),211–217.

Bex, F., Green, P., Marzolf, J. et al. (2003)The human LRP5 G171V mutation inmice alters the skeleton response to limbunloading but not to ovariectomy.Journal of Bone and Mineral Research 18(suppl. 2), S463.

Bikle, D.D., Sakata, T., Leary, C. et al. (2002)Insulin-like growth factor I is requiredfor the anabolic actions of parathyroidhormone on mouse bone. Journal of Boneand Mineral Research 17(9), 1570–1578.

Blum, M., Harris, S.S., Must, A. et al. (2003)Leptin, body composition and bone

mineral density in premenopausalwomen. Calcified Tissue International73(1), 27–32.

Boonen, S., Rosen, C., Bouillon, R. et al.(2002) Musculoskeletal effects of therecombinant human IGF-I/IGF bindingprotein-3 complex in osteoporoticpatients with proximal femoral fracture:a double-blind, placebo-controlled pilotstudy. Journal of Clinical Endocrinologyand Metabolism 87(4), 1593–1599.

Bouxsein, M.L. & Marcus, R. (1994)Overview of exercise and bone mass.Rheumatic Diseases Clinics of NorthAmerica 20(3), 787–802.

Boyden, L.M., Mao, J., Belsky, J. et al. (2002)High bone density due to a mutation inLDL-receptor-related protein 5. NewEngland Journal of Medicine 346(20),1513–1521.

Chappard, D., Minaire, P., Privat, C. et al.(1995) Effects of tiludronate on bone lossin paraplegic patients. Journal of Bone andMineral Research 10(1), 112–118.

Chow, J.W., Fox, S., Jagger, C.J. &Chambers, T.J. (1998) Role forparathyroid hormone in mechanicalresponsiveness of rat bone. AmericanJournal of Physiology 274(1 Pt. 1),E146–E154.

Chrousos, G.P. & Gold, P.W. (1992) Theconcepts of stress and stress systemdisorders. Overview of physical and

Page 440: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

424 chapter 28

behavioral homeostasis. JAMA 267(9),1244–1252.

Cock, T.A. & Auwerx, J. (2003) Leptin:cutting the fat off the bone. Lancet362(9395), 1572–1574.

Cooper, C.S., Taaffe, D.R., Guido, D. et al. (1998) Relationship of chronicendurance exercise to the somatotropicand sex hormone status of older men.European Journal of Endocrinology 138(5),517–523.

Cummings, S.R., Karpf, D.B., Harris, F. et al. (2002) Improvement in spine bonedensity and reduction in risk of vertebralfractures during treatment withantiresorptive drugs. American Journal ofMedicine 112(4), 281–289.

Darmaun, D., Matthews, D.E. & Bier, D.M.(1988) Physiological hypercortisolemiaincreases proteolysis, glutamine, andalanine production. American Journal ofPhysiology 255(3 Pt. 1), E366–E373.

DiPietro, L. & Stachenfeld, N.S. (1997) Thefemale athlete triad. Medicine and Sciencein Sports and Exercise 29(12), 1669–1671.

Drinkwater, B.L., Nilson, K., Chesnut,C.H., 3rd et al. (1984) Bone mineralcontent of amenorrheic andeumenorrheic athletes. New EnglandJournal of Medicine 311(5), 277–281.

Duan, Y., Seeman, E. & Turner, C.H. (2001)The biomechanical basis of vertebralbody fragility in men and women.Journal of Bone and Mineral Research16(12), 2276–2283.

Dueck, C.A., Matt, K.S., Manore, M.M. &Skinner, J.S. (1996) Treatment of athleticamenorrhea with a diet and trainingintervention program. InternationalJournal of Sport Nutrition 6(1), 24–40.

Elefteriou, F., Takeda, S., Liu, X.,Armstrong, D. & Karsenty, G. (2003)Monosodium glutamate-sensitivehypothalamic neurons contribute to thecontrol of bone mass. Endocrinology144(9), 3842–3847.

Forwood, M.R. (1996) Inducible cyclo-oxygenase (COX-2) mediates theinduction of bone formation bymechanical loading in vivo. Journal of Bone and Mineral Research 11(11),1688–1693.

Forwood, M.R., Li, L., Kelly, W.L. &Bennett, M.B. (2001) Growth hormone ispermissive for skeletal adaptation tomechanical loading. Journal of Bone andMineral Research 16(12), 2284–2290.

Fuchs, R.K., Bauer, J.J. & Snow, C.M. (2001)Jumping improves hip and lumbar spinebone mass in prepubescent children: a randomized controlled trial. Journal of Bone and Mineral Research 16(1),148–156.

growth factor-I. Metabolism44(10 Suppl 4), 50–57.

Klein-Nulend, J., Burger, E.H., Semeins,C.M., Raisz, L.G. & Pilbeam, C.C. (1997) Pulsating fluid flow stimulatesprostaglandin release and inducibleprostaglandin G/H synthase mRNAexpression in primary mouse bone cells.Journal of Bone and Mineral Research 12(1),45–51.

Klibanski, A., Biller, B.M., Schoenfeld,D.A., Herzog, D.B. & Saxe, V.C. (1995)The effects of estrogen administration on trabecular bone loss in young womenwith anorexia nervosa. Journal of ClinicalEndocrinology and Metabolism 80(3),898–904.

Kohrt, W.M., Ehsani, A.A. & Birge, S.J., Jr.(1997) Effects of exercise involvingpredominantly either joint-reaction orground-reaction forces on bone mineraldensity in older women. Journal of Bone and Mineral Research 12(8),1253–1261.

Kohrt, W.M., Ehsani, A.A. & Birge, S.J., Jr.(1998) HRT preserves increases in bonemineral density and reductions in bodyfat after a supervised exercise program.Journal of Applied Physiology 84(5),1506–1512.

Lacey, D.L., Timms, E., Tan, H.L. et al.(1998) Osteoprotegerin ligand is acytokine that regulates osteoclastdifferentiation and activation. Cell 93(2),165–176.

Laketic-Ljubojevic, I., Suva, L.J., Maathuis,F.J., Sanders, D. & Skerry, T.M. (1999)Functional characterization of N-methyl-d-aspartic acid-gated channelsin bone cells. Bone 25(6), 631–637.

Lean, J.M., Mackay, A.G., Chow, J.W. & Chambers, T.J. (1996) Osteocyticexpression of mRNA for c-fos and IGF-I:an immediate early gene response to anosteogenic stimulus. American Journal ofPhysiology 270(6 Pt. 1), E937–E945.

Little, R.D., Carulli, J.P., Del Mastro, R.G. et al. (2002) A mutation in the LDLreceptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. American Journal of HumanGenetics 70(1), 11–19.

Loucks, A.B. & Callister, R. (1993)Induction and prevention of low-T3 syndrome in exercising women.American Journal of Physiology 264(5 Pt. 2), R924–R930.

Loucks, A.B., Laughlin, G.A., Mortola, J.F.et al. (1992) Hypothalamic–pituitary–thyroidal function in eumenorrheic andamenorrheic athletes. Journal of ClinicalEndocrinology and Metabolism 75(2),514–518.

Gong, Y., Slee, R.B., Fukai, N. et al. (2001) LDL receptor-related protein 5 (LRP5) affects bone accrual and eyedevelopment. Cell 107(4), 513–523.

Gordon, C.M., Grace, E., Emans, S.J. et al. (2002) Effects of oraldehydroepiandrosterone on bonedensity in young women with anorexianervosa: a randomized trial. Journal ofClinical Endocrinology and Metabolism87(11), 4935–4941.

Grimston, S.K. (1993) An application ofmechanostat theory to research design: a theoretical model. Medicine and Science in Sports and Exercise 25(11),1293–1297.

Grimston, S.K., Willows, N.D. & Hanley,D.A. (1993) Mechanical loading regimeand its relationship to bone mineraldensity in children. Medicine and Sciencein Sports and Exercise 25(11), 1203–1210.

Grinspoon, S., Miller, K., Herzog, D.,Clemmons, D. & Klibanski, A. (2003)Effects of recombinant human insulin-like growth factor (IGF)-I and estrogenadministration on IGF-I, IGF bindingprotein (IGFBP)-2, and IGFBP-3 inanorexia nervosa: a randomized-controlled study. Journal of ClinicalEndocrinology and Metabolism 88(3),1142–1149.

Heaney, R.P., McCarron, D.A., Dawson-Hughes, B. et al. (1999) Dietary changesfavorably affect bone remodeling inolder adults. Journal of American DieteticAssociation 99(10), 1228–1233.

Jones, B.H., Harris, J.M., Vinh, T.N. &Rubin, C. (1989) Exercise-induced stress fractures and stress reactions ofbone: epidemiology, etiology, andclassification. Exercise and Sport SciencesReview 17, 379–422.

Kanders, B., Dempster, D.W. & Lindsay, R.(1988) Interaction of calcium nutritionand physical activity on bone mass inyoung women. Journal of Bone andMineral Research 3(2), 145–149.

Kannus, P., Haapasalo, H., Sankelo, M. et al. (1995) Effect of starting age ofphysical activity on bone mass in thedominant arm of tennis and squashplayers. Annals of Internal Medicine123(1), 27–31.

Kato, M., Patel, M.S., Levasseur, R. et al.(2002) Cbfa1-independent decrease inosteoblast proliferation, osteopenia, and persistent embryonic eyevascularization in mice deficient in Lrp5, a Wnt coreceptor. Journal of CellBiology 157(2), 303–314.

Ketelslegers, J.M., Maiter, D., Maes, M.,Underwood, L.E. & Thissen, J.P. (1995)Nutritional regulation of insulin-like

Page 441: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormone and exercise-induced modulation 425

Marcus, R., Cann, C., Madvig, P. et al.(1985) Menstrual function and bonemass in elite women distance runners.Endocrine and metabolic features.Annals of Internal Medicine 102(2),158–163.

Martin, T.J. & Ng, K.W. (1994) Mechanismsby which cells of the osteoblast lineagecontrol osteoclast formation andactivity. Journal of Cellular Biochemistry56(3), 357–366.

Mason, D.J., Suva, L.J., Genever, P.G. et al. (1997) Mechanically regulatedexpression of a neural glutamatetransporter in bone: a role for excitatoryamino acids as osteotropic agents? Bone20(3), 199–205.

Morris, F.L., Naughton, G.A., Gibbs, J.L.,Carlson, J.S. & Wark, J.D. (1997)Prospective 10-month exerciseintervention in premenarcheal girls:positive effects on bone and lean mass.Journal of Bone and Mineral Research 12(9),1453–1462.

Musey, V.C., Goldstein, S., Farmer, P.K.,Moore, P.B. & Phillips, L.S. (1993)Differential regulation of IGF-1 and IGF-binding protein-1 by dietarycomposition in humans. AmericanJournal of the Medical Sciences 305(3),131–138.

Nakchbandi, I.A., Mitnick, M.A., Lang, R.et al. (2002) Circulating levels ofinterleukin-6 soluble receptor predictrates of bone loss in patients withprimary hyperparathyroidism. Journal of Clinical Endocrinology and Metabolism87(11), 4946–4951.

Neer, R.M., Arnaud, C.D., Zanchetta, J.R. et al. (2001) Effect of parathyroidhormone (1–34) on fractures and bonemineral density in postmenopausalwomen with osteoporosis. New EnglandJournal of Medicine 344(19), 1434–1441.

Nelson, D.A., Barondess, D.A., Hendrix,S.L. & Beck, T.J. (2000) Cross-sectionalgeometry, bone strength, and bone massin the proximal femur in black and whitepostmenopausal women. Journal of Boneand Mineral Research 15(10), 1992–1997.

Neuman, W.F. (1970) Calcium metabolismin space flight. Life Sciences and SpaceResearch 8, 309–315.

Noble, B.S. & Reeve, J. (2000) Osteocytefunction, osteocyte death and bonefracture resistance. Molecular and CellularEndocrinology 159(1–2), 7–13.

Otis, C.L., Drinkwater, B., Johnson, M.,Loucks, A. & Wilmore, J. (1997)American College of Sports Medicineposition stand. The female athlete triad.Medicine and Science in Sports and Exercise29(5), i–ix.

Physiology and Occupational Physiology67(4), 330–334.

Specker, B.L. (1996) Evidence for aninteraction between calcium intake andphysical activity on changes in bonemineral density. Journal of Bone andMineral Research 11(10), 1539–1544.

Srinivasan, S. & Gross, T. (2000)Intermittent rest enhances osteoblasticactivation induced by mechanicalloading. Transactions of the OrthopedicResearch Society 25, 628.

Takeda, S., Elefteriou, F., Levasseur, R. et al. (2002) Leptin regulates boneformation via the sympathetic nervoussystem. Cell 111(3), 305–317.

Thissen, J.P., Ketelslegers, J.M. &Underwood, L.E. (1994) Nutritionalregulation of the insulin-like growthfactors. Endocrine Reviews 15(1), 80–101.

Tomten, S.E., Falch, J.A., Birkeland, K.I.,Hemmersbach, P. & Hostmark, A.T.(1998) Bone mineral density andmenstrual irregularities. A comparativestudy on cortical and trabecular bonestructures in runners with allegednormal eating behavior. InternationalJournal of Sports Medicine 19(2), 92–97.

Turner, C.H. (1999) Site-specific skeletaleffects of exercise: importance ofinterstitial fluid pressure. Bone 24(3),161–162.

Turner, C.H., Takano, Y., Owan, I. &Murrell, G.A. (1996) Nitric oxideinhibitor L-NAME suppressesmechanically induced bone formation in rats. American Journal of Physiology270(4 Pt. 1), E634–E639.

Turner, C.H., Anne, V. & Pidaparti, R.M.(1997) A uniform strain criterion for trabecular bone adaptation: docontinuum-level strain gradients driveadaptation? Journal of Biomechanics 30(6),555–563.

Webster, J.I., Tonelli, L. & Sternberg, E.M.(2002) Neuroendocrine regulation ofimmunity. Annual Review of Immunology20, 125–163.

Wheeler, G.D., Wall, S.R., Belcastro, A.N.& Cumming, D.C. (1984) Reducedserum testosterone and prolactin levelsin male distance runners. JAMA 252(4),514–516.

Williams, N.I., Young, J.C., McArthur, J.W.et al. (1995) Strenuous exercise withcaloric restriction: effect on luteinizinghormone secretion. Medicine and Sciencein Sports and Exercise 27(10), 1390–1398.

Witzke, K.A. & Snow, C.M. (2000) Effectsof plyometric jump training on bonemass in adolescent girls. Medicine andScience in Sports and Exercise 32(6),1051–1057.

Pruitt, L.A., Jackson, R.D., Bartels, R.L. &Lehnhard, H.J. (1992) Weight-trainingeffects on bone mineral density in earlypostmenopausal women. Journal of Boneand Mineral Research 7(2), 179–185.

Qin, Y.X., Kaplan, T., Saldanha, A. &Rubin, C. (2003) Fluid pressuregradients, arising from oscillations inintramedullary pressure, is correlatedwith the formation of bone andinhibition of intracortical porosity.Journal of Biomechanics 36(10), 1427–1437.

Robey, P.B. & Bianco, P. (1999) Cellularmechanism of age related bone loss. In:Aging Skeleton (Rosen, C.J., Glowacki, J.& Bilezikian, J., eds.). Academic Press,San Diego: 145–157.

Rosen, C.J. (2003) Restoring aging bones.Scientific American 288(3), 70–77.

Rosen, C.J. & Donahue, L.R. (1998) Insulin-like growth factors and bone: the osteoporosis connection revisited.Proceedings of the Society for ExperimentalBiology and Medicine 219(1), 1–7.

Rosen, C.J. & Kessenich, C. (1996) The roleof IGF-I in senesence: implications fortherapy. Endocrinologists 6, 102–106.

Rubin, C.T. & Lanyon, L.E. (1984)Regulation of bone formation by applieddynamic loads. Journal of Bone and JointSurgery. American Volume 66(3), 397–402.

Rubin, C.T. & Lanyon, L.E. (1985)Regulation of bone mass by mechanicalstrain magnitude. Calcified TissueInternational 37(4), 411–417.

Rubin, J., Murphy, T.C., Zhu, L. et al. (2003) Mechanical strain differentiallyregulates eNOS and RANKL expressionvia ERK1/2 in primary bone stromalcells. Journal of Bone and Mineral Research18 (suppl. 2), S10.

Sakata, T., Halloran, B.P., Elalieh, H.Z. et al. (2003) Skeletal unloading inducesresistance to insulin-like growth factor Ion bone formation. Bone 32(6), 669–680.

Sato, Y., Kaji, M., Higuchi, F. et al. (2001) Changes in bone and calciummetabolism following hip fracture in elderly patients. OsteoporosisInternational 12(6), 445–449.

Schurch, M.A., Rizzoli, R., Slosman, D. et al. (1998) Protein supplementsincrease serum insulin-like growthfactor-I levels and attenuate proximalfemur bone loss in patients with recenthip fracture. A randomized, double-blind, placebo-controlled trial. Annals ofInternal Medicine 128(10), 801–809.

Smith, R. & Rutherford, O.M. (1993) Spineand total body bone mineral density and serum testosterone levels in maleathletes. European Journal of Applied

Page 442: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

426

Introduction

In order to sustain life, there must be a continuoussupply of energy obtained from the diet. The prim-ary energy-providing nutrients are protein, carbo-hydrate and fat. When food is ingested, there is anacute increase in plasma metabolites and hormonesthat is dependent, in part, on timing and macro-nutrient distribution of the meal. Nutrients and hormones then interact at target tissues to regulatecellular processes. In the context of body composi-tion, nutrients and hormones have important rolesin regulating skeletal muscle protein and adiposetriacylglycerol balance, which over time impactsbody composition. Exercise also has independenteffects on nutrients and hormone availability andinteracts with feeding to create a unique setting that impacts metabolic processes including bodycomposition. This general scenario is depicted inFig. 29.1. The postprandial phase of metabolismincludes the time when nutrients are being absorbedfrom the gut and appear in the circulation. This timeperiod continues several hours after feeding andtherefore most people are in a postprandial phasefor the majority of their life. Similar to the situationlinking the postprandial lipoprotein response to car-diovascular disease (Patsch et al. 1992; Ebenbichleret al. 1995), the postprandial hormonal responsecould be viewed as more physiologically importantthan the postabsorptive hormonal environment,particularly for enhancing body composition.

Given the important regulatory affects of hor-mones, it is surprising that a more systematic andcomprehensive study of the effects of hormonal

responses to feeding has not been undertaken. Thestudies reviewed in this chapter are from diverseperspectives but share the common design com-ponent of measuring the hormonal response to foodintake. We synthesized these postprandial hor-monal data in an attempt to shed light on the con-sistency or lack thereof related to the hormonalresponse to intake of meals with different com-position with and without exercise. We discuss separately the effects of meals on growth hormone(GH), insulin-like growth factor I (IGF-I), testoster-one, cortisol and insulin because these hormoneshave a major role in regulating protein and lipidmetabolism. For each hormone, we overview thepostprandial response to meals of different macro-nutrient distribution and the exercise-induced hor-monal response to feeding before, during, or afterexercise. Although theoretical, the implications ofdiet-induced hormonal responses on body composi-tion are discussed. We focus primarily on humanstudies and only include animal work to providesupporting information.

Effects of diet on hormones

Growth hormone

GH is a peptide hormone secreted from the anter-ior pituitary. In skeletal muscle, GH promotes a positive protein balance by increasing protein syn-thesis and possibly inhibiting protein breakdown(Rooyackers & Nair 1997). These effects are contro-versial in part because it is difficult to separate outthe effects mediated through stimulation of hepatic

Chapter 29

Diet and Hormonal Responses: Potential Impact onBody Composition

JEFF S. VOLEK AND MATTHEW J. SHARMAN

Page 443: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 427

or skeletal muscle IGF-I. In adipose tissue, GHincreases lipolysis and there has been specific inter-est in the C-terminal fragment of GH shown to havemarked lipolytic and antilipogenic activity in vitroand when administered to animals (Heffernan et al.2000). GH dramatically decreases lipogenesis with aconcomitant increase in muscle mass indicating apowerful nutrient partitioning effect (Etherton 2000).

postprandial response to feeding

The GH response to meals has been shown to bequite variable (Baker et al. 1972; van Loon et al. 2003),which may be explained in part by the pulsatile

release pattern of GH. Studies have shown that pro-tein, fat and carbohydrate each have independenteffects on regulation of GH secretion. An oral gluc-ose tolerance test results in significantly reducedGH levels (Hjalmarsen et al. 1996; Bernardi et al.1999; Nakagawa et al. 2002). Carbohydrate intake(0.7 g·kg–1·h–1) alone or in combination with protein(0.35 g·kg–1·h–1) resulted in declining GH levels overa 2-h period (van Loon et al. 2003). An oral glucosetolerance test (75 g glucose) resulted in a slight andgradual decrease in GH for 2 h followed by asignificant increase that peaked at 240 min (Frystyket al. 1997). Thus, hyperglycemia is associated with a decrease in GH, which may be followed by

Skeletalmuscle

Bodycomposition

Adipocyte

Exercise stimulusProteinCarbohydrate

Fat

Glucose GH

IGF-I

Testosterone

Cortisol

InsulinChylomicrons

Amino acids

Intake ofmacronutrients

Alterationin plasmahormones

Alterationin plasma

metabolites

Nutrients and hormones interactat target tissues to regulate

protein and lipid balance

FATAG

AAPRO

Fig. 29.1 Intake of macronutrients results in appearance of glucose, amino acids and triacylglycerols in the form ofchylomicrons in the plasma. These nutrients are also affected by exercise and together contribute to a release of hormonesthat then partition nutrients and interact with target tissues such as skeletal muscle and adipose tissue to regulate amongother processes, protein synthesis/breakdown and adipose tissue lipolysis/lipogenesis, the balance of which over timeimpacts body composition. AA, amino acids; FA, fatty acids; GH, growth hormone; IGF-I, insulin-like growth factor-I;PRO, protein; TAG, triacylglycerols.

Page 444: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

428 chapter 29

rebound hypoglycemia and a subsequent increasein GH. This is consistent with work showing hypo-glycemia is a potent stimulator of GH (Roth et al.1963). Thus in response to carbohydrate, GH tendsto be decreased and increased in the early and latepostprandial periods, respectively.

Although infusion and, in some cases, oral inges-tion of large doses of certain amino acids (e.g. arginine, lysine and ornithine) can increase GH levels, the effect is quite variable and reduced byphysical training and high-protein diets (reviewedby Chromiak & Antonio 2002). Ingestion of beefsteak resulted in an increase in GH, presumably dueto the amino acid content, but the increase was pre-vented when heparin was injected to acutely elevatecirculating fatty acids (Fineberg et al. 1972). In linewith an inhibitory effect of fatty acids on GH, otherwork has confirmed that circulating fatty acids, butnot triacylglycerols, inhibit GH secretion (Blackardet al. 1971). The time course of the GH response tofood is not entirely clear. Acute ingestion of a liquidsupplement (520 kcal [2177 kJ]) rich in fat or rich incarbohydrate did not affect GH measured 35 minlater (Cappon et al. 1993) nor did a mixed carbo-hydrate and protein meal (216 kcal [903 kJ]) affectGH levels measured 90 min later (Carli et al. 1992).Alcohol ingestion (0.45 g ethanol·kg–1) consumedeach hour for 3 h had no effect on GH responsescompared to water (Rojdmark et al. 2000).

exercise-induced responses to feeding

Carbohydrate and protein intake alter the GHresponse to resistance exercise. A protein and car-bohydrate supplement consumed immediately and 2 h after resistance exercise increased GH duringlate recovery when glucose levels were lower com-pared to a placebo (Chandler et al. 1994), consistentwith the known effects of hypoglycemia on GH. Our laboratory reported that a protein and carbohy-drate supplement consumed before and immedi-ately after a bout of resistance exercise resulted in anenhanced acute GH response from 0–30 min post-exercise compared to a non-caloric placebo despitesimilar glucose levels between trials (Kraemer et al.1998). Another study reported that protein and car-bohydrate intake after resistance exercise had no

significant effect on GH responses (Williams et al.2002).

Meals also alter the GH response to submaximalexercise. Whitley et al. (1998) compared the effects offasting to isoenergetic (956 kcal [40031 kJ]) high-fat(74% fat) and high-carbohydrate (86% carbohy-drate) meals consumed 4 h prior to cycling for 90 minon GH responses. The GH response during the fast-ing and carbohydrate trial were similar and signific-antly higher compared to the high-fat trial. Consistentwith the effects of glucose and fatty acids on GHsecretion, the lower GH levels during exercise afterthe fat-rich meal was associated with higher bloodglucose and fatty acid levels. Cappon et al. (1993) com-pared the effects of isoenergetic (520 kcal [2177 kJ])high-fat and high-carbohydrate liquid meals con-sumed 45 min before 10 min of intense exercise onpost-exercise GH responses. Compared to ingestionof a noncaloric placebo, the post-exercise GH areaunder the curve was decreased by –54% after thehigh-fat meal and by –25% after the high-carbohy-drate meal. Post-exercise somatostatin was elevatedafter the high-fat meal and the authors postulate a potential link between dietary fat, somatostatinand GH (Cappon et al. 1993). Somatostatin alsodecreases ghrelin (Schaller et al. 2003), a recentlyidentified potent GH-releasing peptide primarilyproduced in the stomach, providing an alternativefat-induced mechanism to decrease GH.

The finding that a high-fat meal reduces post-exercise GH more than a high-carbohydrate meal is somewhat counterintuitive since carbohydratefeeding would likely lead to higher glucose levels,which should inhibit GH. In line with this role of GH as a counter-regulatory hormone, severalstudies have shown that compared to placebo, car-bohydrate beverages consumed before, during andafter exercise result in higher blood glucose levelsand lower GH responses to exercise (Bonen et al.1980; Tsintzas et al. 1996; Utter et al. 1999). Miller et al.(2002) showed that carbohydrate provided during 2 h of cycling blunted the post-exercise GH responsecompared to non-fat milk and a non-caloric placebodespite similar glucose levels among the three trialssuggesting that GH response to exercise are medi-ated by factors other than blood glucose. Studieshave also shown that carbohydrate provided before

Page 445: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 429

and during 2 h of cycling (Murray et al. 1995) or rowing (Henson et al. 2000) do not affect GH levelsdespite differences in glucose levels. In one of thesestudies (Murray et al. 1995), subjects also consumedthe beverages with and without nicotinic acid, aninhibitor of lipolysis. Nicotinic acid consumed witheither water or carbohydrate prevented the increasein fatty acids and resulted in increased post-exerciseGH levels compared to water and carbohydratewithout nicotinic acid (Murray et al. 1995). Thus, theGH response to exercise appears to be dependent inpart on glucose levels, but fatty acid levels also exertan independent effect.

Exercise-induced elevations in GH to a high-fatdiet or fasting are accompanied by a more rapiddecline in plasma insulin and glucose concentra-tions during exercise suggesting that glucose sensit-ive receptors may modulate the GH response toexercise (Galbo et al. 1979). Glucose infusion at theend of exercise does not attenuate the greater GHresponse observed after a fat-rich diet (Galbo et al.1979; Johannessen et al. 1981) again suggesting thatother substrates (e.g. glycogen, ketones, fatty acids)or hormones (e.g. insulin, catecholamines) may beinvolved in the regulation of GH secretion.

Insulin-like growth factor I

IGF-I is an anabolic hormone that stimulates growthin almost all tissues and is likely responsible formany of the effects of GH. IGF-I is primarily pro-duced in the liver but also in other tissues includingskeletal muscle under stimulation by GH. In skeletalmuscle, IGF-I increases protein balance primarily byincreasing protein synthesis (Rooyackers & Nair1997). In adipose tissue, IGF-I has insulin-like effectsstimulating glucose uptake and inhibiting lipolysis(Siddals et al. 2002).

postprandial response to feeding

Effects of macronutrients on total IGF-I levels. An oralglucose tolerance test (75 g glucose) had no effect ontotal IGF-I levels measured every 30 min for 330min in healthy subjects (Frystyk et al. 1997). Carbo-hydrate intake (0.7 g·kg–1·h–1) alone or in combina-tion with protein (0.35 g·kg–1·h–1) had no effect on

total IGF-I levels measured 2 h later (van Loon et al.2003). In another study, total IGF-I levels were notaffected when measured 4 h after a carbohydrate-rich breakfast (Bereket et al. 1996). A mixed mealhad no effect on total IGF-I levels measured over thenext 5 h (Svanberg et al. 2000) and another studyshowed no changes in total IGF-I levels over a 24-hperiod during which subjects were fed mixed meals(Frystyk et al. 2003). Consumption of ethanol didreduce IGF-I levels during the late postprandialperiod (5–7 h after ingestion) (Rojdmark et al. 2000).Thus, feeding does not appear to affect total IGF-Ilevels with the exception of a delayed decrease afteralcohol ingestion. However, feeding does appear toalter IGF-I binding proteins.

Effects of macronutrients on other components of theIGF-I system. There are six high affinity insulin-likegrowth factor binding proteins (IGFBP-1 to 6) thatcirculate bound to the vast majority of IGF-I. IGFBP-1 shows the most rapid dynamic regulation inplasma in response to meals and it has been sug-gested that it contributes to glucose regulation by virtue of its role in countering the insulin-likebioactivity of IGF-I (i.e. hypoglycemic effect), mostlikely by controlling free levels of IGF-I (Lee et al.1993, 1997). After an oral glucose tolerance test,IGFBP-1 gradually decreased reaching a level –52%below baseline after 180 min followed by a gradualincrease to values 74% above baseline at 5 h (Frystyket al. 1997). Opposite to IGFBP-1, free IGF-I wassignificantly decreased (–29% to –38%) during thelate postprandial period (270–330 min) and wasinversely related to IGFBP-1 at baseline and duringthe late postprandial period. Other studies haveobserved decreases in IGFBP-1 in response to mixedmeals (Bereket et al. 1996; Frystyk et al. 2003), oralglucose (Bernardi et al. 1999) or intravenous glucoseand insulin injections (Nyomba et al. 1997). Inverseassociations have been shown between IGFBP-1 andglucose, insulin and cortisol levels (Hopkins et al.1994; Holden et al. 1995; Frystyk et al. 1997; Ricart &Fernandez-Real 2001). Acute ingestion of ethanolresulted in a rapid rise in IGFBP-1 that was independ-ent of changes in glucose, insulin and GH, suggest-ing its regulation is partially mediated by othermechanisms.

Page 446: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

430 chapter 29

Although there is a significant amount of evid-ence supporting the role of the GH–IGF-I axis in glucose metabolism (Holt et al. 2003), there has beendebate whether the IGF-I system is important in glucose regulation in response to metabolic stressors(e.g. feeding and/or exercise). This is because freeIGF-I levels are generally not affected during theearly postprandial period when IGFBP-1, glucoseand insulin are elevated but free IGF-I levels arereduced during the late postprandial period andduring the night when glucose and insulin are normalized (Frystyk et al. 1997, 2003). Thus, themajority of evidence indicates that the IGF-I systemdoes not partake in meal-related glucose regulationbut does contribute to glucose disposal during thepost-absorptive state.

Since IGF-I is produced in many tissues includingskeletal muscle, it is possible that the IGF-I system is contributing to meal-related glucose regulationthrough an autocrine and/or paracrine mechanism.However, this hypothesis is unlikely based onfindings from a recent study showing that feedingdoes not affect postprandial skeletal muscle IGF-ImRNA expression in humans (Svanberg et al. 2000).

exercise-induced responses to feeding

Effects of macronutrients on total exercise-induced IGF-Iresponses. Consistent with the lack of a change offood on total IGF-I levels, studies have shown thatpre-exercise and post-exercise meals do not alter thetotal IGF-I response to exercise (Cappon et al. 1994;Hopkins et al. 1994; Kraemer et al. 1998; Anthony et al. 2001). A pre-exercise carbohydrate- and fat-richmeal had the same effect as a non-caloric placebo on IGF-I responses to 10 min of intense cycling exercise, suggesting that pre-exercise feeding andmeal composition do not influence exercise-inducedIGF-I levels (Cappon et al. 1994). There were no differences in total IGF-I levels in a group of men feda placebo or a glucose polymer solution duringcycling exercise to fatigue (Hopkins et al. 1994). Ourlaboratory showed that a protein and carbohydratesupplement consumed 2 h before and immediatelyafter a bout of whole body resistance exercise did not alter the exercise-induced IGF-I response(Kraemer et al. 1998). Finally, Anthony et al. (2001)

showed that a nutritional complete meal fed imme-diately after 2 h of treadmill running had no effecton exercised induced total IGF-I levels compared toa food-deprived group of rats. Thus, acute feedingof any composition does not appear to affect sys-temic levels of total IGF-I.

Effects of macronutrients on exercise-induced compon-ents of the IGF-I system. Similar to the situation afterfeeding, it has been proposed that IGFBP-1 mayhave an important role in glucose regulation duringand after exercise (Koistinen et al. 1996). Comparedto placebo, exercise-induced IGFBP-1 levels werereduced in response to feeding of carbohydrate(Hopkins et al. 1994). In this study, IGFBP-1 and gluc-ose levels were correlated in the control conditionbut not during the carbohydrate feeding trial, suggesting that other factors other than glucose andinsulin regulate IGFBP-1 responses to prolongedexercise (Hopkins et al. 1994). Anthony et al. (2001)showed that provision of a nutritionally completemeal immediately after exercise did not alter theexercise-induced increase in hepatic IGFBP-1 mRNAexpression or circulating IGFBP-1 levels despitehigher glucose and insulin levels. Again, this is con-sistent with the hypothesis that exercise-inducedincreases in IGFBP-1 levels are not directly regu-lated by circulating glucose levels and do not assist in preventing IGF-I induced hypoglycemia by binding free IGF-I in plasma. Findings from a re-cent study showed high correlations between liverglycogen and IGFBP-1 responses to exercise, sug-gesting that the magnitude of liver glycogen deple-tion may mediate IGFBP-1 responses to exercise(Lavoie et al. 2002). Although speculative, it is pos-sible that IGFBP-1 responses to exercise are modulat-ing anabolic (growth), as opposed to the metabolic(glucose-lowering), effects of IGF-I.

Testosterone

Testosterone is a steroid hormone secreted fromspecialized cells in the testes in men and from theovaries in women. In skeletal muscle, testosteroneincreases protein balance primarily by increasingprotein synthesis whereas the effects on proteinbreakdown are unclear (Rooyackers & Nair 1997).

Page 447: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 431

In adipose tissue, testosterone inhibits lipid up-take and LPL activity, and stimulates lipolysis byincreasing the number of lipolytic β-adrenergicreceptors (De Pergola 2000).

postprandial response to feeding

The testosterone response to isocaloric (800 kcal[3350 kJ]) meals that were high-fat (57% fat, 9% pro-tein, 34% carbohydrate) or low-fat (1% fat, 26% pro-tein, 73% carbohydrate) was examined in healthymen (Meikle et al. 1990). Testosterone was not affectedafter the low-fat meal, but postprandial total andfree testosterone concentrations were approxim-ately 30% lower for 4 h after the high-fat meal. Thefat-induced decrease was not related to changes in other steroids (i.e. estrone, estradiol, dihydro-testosterone, lutenizing hormone [LH], percent free testosterone, or sex hormone binding globulin[SHBG] binding capacity). This study indicates fat-rich meals, but not carbohydrate-rich meals, decreasepostprandial testosterone levels. Our laboratory hasalso observed a significant reduction in total testo-sterone (–22%) and free testosterone (–23%) after afat-rich meal in healthy men (Volek et al. 2001).

Another study compared testosterone responsesto meals with different sources of protein (soy versus meat), amounts of fat (lean versus fatty meat)and sources of fat (animal versus vegetable) (Habitoet al. 2001). The decrease in testosterone was greaterafter a low-fat meal consisting of lean meat (–22%)compared to tofu (–15%); a meal consisting of lean meat (–22%) compared to lean meat cooked withanimal fat meal (–9%); and a meal consisting of leanmeat cooked with vegetable fat (–17%) compared tovegetable oil (–9%). Although these results do notnecessarily agree with the findings mentionedabove that only fat-rich meals reduce testosterone,the study does provide further evidence that thecomposition of meals, particularly the amount andtype of fat, influences the circulating testoster-one response to meals. All the meals resulted indecreases in testosterone, increases in LH and nochanges in SHBG.

A number of studies have examined the testo-sterone response to an oral glucose tolerance test.The majority of these studies have been in women,

with only one to our knowledge performed in men(Hjalmarsen et al. 1996). In this study, total testo-sterone and estimated free testosterone levels weresignificantly decreased during a 2-h oral glucose to-lerance test. There were no changes in SHBG but LHwas significantly increased (Hjalmarsen et al. 1996).

The above studies have all involved men. Studiesgenerally indicate that testosterone is also decreasedmodestly after an oral glucose tolerance test inhealthy normal-weight women (Smith, S. et al. 1987;Falcone et al. 1990; Tiitinen et al. 1990; Aizawa &Niimura 1996; Ivandic et al. 1999), which may beattributed in part to the normal diurnal variation inthe hormone. There have been a number of studiesexamining the androgen response to oral glucosetolerance tests in women with polycystic ovariansyndrome (PCOS) because they often exhibit bothelevated androgens and insulin, which are associ-ated with insulin resistance. Insulin acts to stimulatetestosterone in the ovaries (Cara & Rosenfield 1988),and thus hyperinsulinemia has been hypothesizedto play a pathogenic role in women with PCOS(Bergh et al. 1993). In hyperandrogenic women orthose with PCOS, testosterone responses to glucosetolerance tests also tend to decrease in a similarmanner as healthy controls (Falcone et al. 1990;Tropeano et al. 1994) unless the women are alsoinsulin resistant (Smith, S. et al. 1987) or obese(Tiitinen et al. 1990), and then there is a slightincrease in testosterone.

In summary, testosterone levels consistently dropafter feeding and there is evidence this is dependenton the composition of the meal, particularly fat content. Insulin may play a role in explaining someof the variability in testosterone responses to mealsbecause in men insulin and testosterone tend toexhibit an inverse relation whereas in women thereis a positive association, especially in women withPCOS (Haffner et al. 1994). However, findings fromstudies using the euglycemic hyperinsulinemicclamp procedure cast doubt on the hypothesis thatinsulin mediates the testosterone response to feed-ing because acute elevations in insulin were shownto have no effect on total or free testosterone levelsin healthy normal-weight men (Ebeling et al. 1995;Pasquali et al. 1997) or women (Diamond et al. 1991).The mechanism by which fat-rich meals decrease

Page 448: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

432 chapter 29

postprandial testosterone concentrations may berelated to the elevated chylomicrons or fatty acidsafter a fat-rich meal because they inhibit LH-stimu-lated testosterone production in isolated Leydigcells (Meikle et al. 1989, 1990). It is possible thatspecific nutrients may interact with regions of thetestes and impact on the modulation of testosterone.Alteration in the testicular plasma membrane andchanges in the responsiveness of Leydig cells andsubsequent testosterone synthesis as a result ofingestion of different compositions of lipids hasbeen reported in rats (Sebokova et al. 1988, 1990).

exercise-induced responses to feeding

Resistance exercise has been shown to result inacute elevations of testosterone that peak early after exercise and return to baseline by about 60 minpost-exercise; pre- and post-exercise meals alter this response (Kraemer et al. 1998). Our laboratoryshowed that a protein and carbohydrate beverageconsumed 2 h before and immediately after a boutof whole body resistance exercise resulted in anincrease in testosterone immediately after exercisefollowed by a sharp decrease to values that weresignificantly below baseline (Kraemer et al. 1998). In this study, the exact same exercise and supple-mentation protocol was performed on 3 consecutivedays. The greater reduction in post-exercise testo-sterone with pre- and post-exercise protein and carbohydrate intake compared to placebo wasobserved on all 3 days, emphasizing the consistencyand reproducibility of the response (Kraemer et al.1998). Chandler et al. (1994) examined the post-exercise testosterone response to supplements con-taining either protein alone, carbohydrate alone, ora combination of protein and carbohydrate con-sumed immediately and 2 h after a bout of resist-ance exercise in healthy men. In agreement withfindings from our study, testosterone had decreasedto values below baseline by 30 min post-exerciseduring all the supplement treatments compared toplacebo. Testosterone values remained significantlybelow baseline 5–6 h whereas testosterone returnedto baseline shortly after exercise and stabilizedthroughout the recovery period (Chandler et al.1994). In yet another study, Bloomer et al. (2000)

compared the post-exercise testosterone responsesto a mixed meal, an isocaloric beverage of similarnutrient content, and an isocaloric carbohydratebeverage consumed immediately and 2, 4 and 7 hafter exercise. Compared to a placebo, post-resist-ance exercise testosterone levels were lower during all the meals at 0.5, 2.5, 4.5 and 8 h post-exercise.There is one study that did not show a difference infree testosterone responses to resistance exercisebetween carbohydrate and a water placebo con-sumed during exercise (Tarpenning et al. 2001).

Collectively, these studies indicate that pre- andpost-exercise meals decrease post-exercise testo-sterone levels compared to fasting. The decreasecould be due in part to a decrease in the synthesis/secretion of testosterone and/or an increase inmetabolic clearance. Chandler et al. (1994) showedthat the decrease in post-exercise testosterone wasnot associated with a decrease in LH arguingagainst a decrease in the rate of testosterone secre-tion, however there could still be a decrease in thetesticular responsiveness to LH. Since post-exercisemeals increase muscle-specific protein synthesisduring recovery (Tipton et al. 1999a, 2001; Rasmussenet al. 2000), the lower testosterone levels could bedue in part to increased uptake in active skeletalmuscle. In support of this hypothesis, recent work inour laboratory has shown that the meal-induceddecrease in testosterone after resistance exercise corresponds with an increase in skeletal muscleandrogen receptor content measured 60 min afterexercise (unpublished observations).

Cortisol

Cortisol is an adrenal steroid hormone that is regulated by pituitary adrenocorticotropin (ACTH),which in turn is under the influence of hypothal-amic corticotropin-releasing hormone (CRH). Thishypothalamic–pituitary–adrenal (HPA) axis is sens-itive to a variety of different stressors includingfeeding. At the whole body level, cortisol increasesprotein breakdown but the effects in skeletal muscleare unclear (Rooyackers & Nair 1997). Physiologicalconcentrations of cortisol increase lipolysis in adip-ose tissue but the effects are less potent than GH(Divertie et al. 1991; Djurhuus et al. 2002).

Page 449: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 433

postprandial response to feeding

The effects of feeding on cortisol are complicated bythe fact that cortisol has a distinct diurnal variationand is sensitive to a number of internal and externalstressors. In general, feeding has been shown toincrease cortisol levels and the response is particu-larly evident at the noon meal (Follenius et al. 1982;Knoll et al. 1984), which may serve to synchronizethe diurnal rhythm in HPA axis activity (Leal &Moreira 1997). Although few studies have addressedthe influence of meal composition on the HPA axis,it appears that protein has the greatest stimulatoryeffect on cortisol (Slag et al. 1981; Ishizuka et al. 1983;Gibson et al. 1999) whereas carbohydrate feedinginhibits cortisol (Jezova-Repcekova et al. 1980).

In healthy men, salivary cortisol was shown toincrease substantially after a mid-day protein-richmeal (550 kcal [2299 kJ] with 39% protein) com-pared to fasting and this response although variablefrom subject to subject was highly reproduciblewithin subjects (Gibson et al. 1999). In healthy women,salivary cortisol was also significantly increasedafter a mid-day high-protein (630 kcal [2633 kJ] and32% protein) compared to an isocaloric low-protein(5% protein) meal, which resulted in a decrease in cortisol (Gibson et al. 1999). Other studies haveshown that meals containing 20–40% proteinenhance postprandial cortisol levels compared tomeals with high carbohydrate or fat content andthat protein-free glucose or fat-rich meals do notstimulate cortisol release (Slag et al. 1981; Ishizuka et al. 1983). The stimulation of cortisol by proteinmay be due to specific essential amino acids such astyrosine and tryptophan (Ishizuka et al. 1983).

The findings from the studies mentioned abovethat protein has a stimulatory effect on cortisol butcarbohydrates have a negligible effect appear to bein conflict with other work that has shown glucoseto be important in the regulation of the HPA axis.Gonzalez-Bono et al. (2002) fed subjects either glu-cose, protein, fat or water and then 45 min later wereexposed to a psychosocial stress test. There were nodifferences in cortisol levels measured 45 min afterthe meals, which the authors suggest may have beendue to the timing of the meals. However, cortisollevels after the stress test were significantly higher

after the glucose load compared to all other trialsand the stress-induced cortisol response was posit-ively correlated with glucose changes before thestress test. Since restoration of the stress-inducedHPA response after fasting was limited to the gluc-ose load, which was the only trial to increase bloodglucose, the authors suggest that HPA responsive-ness is under control of centers sensing blood glucose levels, presumably in the hypothalamus(Gonzala-Bono et al. 2002).

Part of the discrepancy between the effects ofmeal composition on postprandial cortisol may beexplained by differences in the pattern of body fatdistribution. Vicennati et al. (2002) recently showedthat women with abdominal fat distribution have a higher cortisol response to a high-carbohydratemeal compared to a high-protein/high-fat meal andthat women with a peripheral fat distributiondemonstrate the opposite effect, which was more inline with the response of a control normal-weightgroup of women.

Postprandial cortisol measurements must beinterpreted in the context of the normal diurnal variation of the hormone. For example, a morningglucose tolerance test results in a decrease in cortisolfor several hours after the meal (Walker et al. 2000;Reynolds et al. 2001, 2003). However, postprandialcortisol values are higher compared to the normaldiurnal drop in cortisol in the morning hours(Reynolds et al. 2001).

The findings above showing a stimulatory effectof glucose on cortisol secretion are consistent withthe findings of Gonzalez-Bono et al. (2002) and thenotion that the HPA axis is responsive to increasesin circulating glucose, but the precise mechanismsregulating this effect remain unresolved. A role ofα1-adrenorecptors modulating ACTH secretion hasbeen proposed to mediate postprandial cortisolresponses (Al-Damluji et al. 1987). In these experi-ments, infusion of methoxamine (an α1-adrenore-ceptor agonist) enhanced and thymoxamine (anα1-adrenoreceptor antagonist) attenuated the ACTHand cortisol response to a standard meal. Theseauthors also showed that patients with a recent pitu-itary ACTH deficiency and normal adrenal glandsshowed no ACTH or cortisol rise after a standardmeal (Al-Damluji et al. 1987). In further support of a

Page 450: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

434 chapter 29

central mechanism, postprandial ACTH and cortisollevels have been shown to be positively correlatedafter a meal (Vicennati et al. 2002). However, not alldata point to a hypothalamic–pituitary mediatedmechanism to explain postprandial cortisol secre-tion (Fehm et al. 1983). In summary, cortisol tends toincrease after feeding, especially protein, but theresponse is dependent on the timing and composi-tion of the meal, fat distribution pattern and must be compared to the normal diurnal variation of thehormone.

exercise-induced responses to feeding

A large number of studies in the last 25 years haveexamined the effects of feeding before, during andafter exercise on cortisol responses. The majority of these have examined carbohydrate feeding and a few have studied how different combinations ofmacronutrients influence cortisol. In terms of stud-ies that measured the cortisol response to resistanceexercise, the majority indicate that intake of carbo-hydrate or carbohydrate combined with proteinbefore and after resistance exercise does not alter the cortisol response compared to placebo (Kraemeret al. 1998; Bloomer et al. 2000; Koch et al. 2001;Williams et al. 2002). One study showed that carbo-hydrate intake during an acute bout of resistanceexercise significantly blunted the cortisol response(Tarpenning et al. 2001). This study further showedthat the reduction in post-resistance exercise cortisolwas significantly related to increases in muscle fiberhypertrophy.

Studies that measured the cortisol response toprolonged endurance or high-intensity exercise aregenerally mixed with several showing that carbohy-drate intake before and during exercise results inlower cortisol responses (Mitchell et al. 1990, 1998;Murray et al. 1991, 1995; Deuster et al. 1992; Niemanet al. 1998, 2003; Utter et al. 1999; Bacurau et al. 2002;Bishop et al. 2002; Green et al. 2003) whereas othershave shown no change in cortisol with carbohydratefeeding (Bonen et al. 1980; Tsintzas et al. 1996; Bishopet al. 1999, 2001; Henson et al. 2000; Miller et al. 2002).

The reduction in the exercise-induced cortisolresponse after carbohydrate supplementation maycontribute to a more favorable immune response to

exercise (Nehlsen-Cannarella et al. 1997; Henson et al. 1998; Nieman 1998). However, the favorableeffects of carbohydrate feeding on immune functionduring and after exercise may not be mediatedthrough a lower cortisol response (Green et al. 2003),but rather some other mechanism such as bettermaintenance of plasma glucose or glutamine levels,an important fuel source for immune cells (Bacurauet al. 2002). A recent study showed that carbohy-drate intake reduced the plasma interleukin-6 (IL-6)response to exercise, but IL-6 mRNA expression in skeletal muscle was not affected suggesting thatcarbohydrate feeding during exercise attenuates IL-6 production by tissues other than skeletal muscle(Starkie et al. 2001).

Insulin

Insulin is a peptide hormone secreted by the pan-creas. In skeletal muscle, insulin has anabolic effectsby increasing amino acid uptake and protein synthe-sis and inhibiting protein breakdown (Rooyackers& Nair 1997). Insulin is generally accepted as a stimu-lator of protein synthesis only when adequateamino acids are available (Kimball & Jefferson 2002;Kimball et al. 2002). Insulin has long been consid-ered the most important regulator of adipose tissuemetabolism. Adipose tissue lipolysis is exquisitelysensitive to insulin at physiological concentrations(Jensen et al. 1989). Small-to-moderate decreases in insulin can increase lipolysis several-fold, theresponse being virtually immediate. Insulin alsostimulates lipogenesis by increasing glucose uptakeand activating lipogenic and glycolytic enzymes(Kersten 2001).

postprandial response to feeding

Carbohydrate ingestion leads to an increase in bloodglucose and a relatively similar increase in insulinconcentrations (Nuttall et al. 1985). A meal rich in fatresults in lower insulin responses compared tomeals rich in either carbohydrate (Nuttall et al. 1985)or protein (Ullrich et al. 1985). Carbohydrate inges-tion results in elevated glucose and insulin levelsthat depend primarily on the glycemic index or,more precisely, the glycemic load of the food. The

Page 451: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 435

glycemic index was developed in 1981 and refers to the effect of standard amounts of individualfoods containing 50 g of available carbohydrate onblood glucose compared with that of a control food(usually white bread or glucose) ( Jenkins et al. 1981).Comprehensive tables describing the glycemicindex of over 1000 foods exist (Foster-Powell et al.2002). To account for differences in the amount ofcarbohydrate in foods or meals, the concept of glycemic load was introduced, which is the pro-duct of glycemic index and carbohydrate amount(Salmeron et al. 1997). The glycemic load can predictglucose and insulin responses to individual foodsacross a wide range of portion sizes (Brand-Miller et al. 2003). Certain amino acids can increase insulinand thus carbohydrate combined with protein canenhance the insulin response (Nuttall et al. 1984,1985).

exercise-induced responses to feeding

Given the synergistic effects of carbohydrate andprotein on insulin responses, there has been someinterest in combining protein with carbohydrate tomaximize insulin secretion in the hopes of enhanc-ing post-exercise glycogen resynthesis (Zawadzki et al. 1992; van Loon et al. 2000a) and proteinanabolism (Rasmussen et al. 2000; Tipton et al. 2001).In a study designed to determine the optimalinsulinotropic mixture of protein and carbohydrate,it was determined that carbohydrate (0.7 g·kg–1·h–1)consisting of 50% glucose and 50% maltodextrincombined with protein (0.35 g·kg–1·h–1) consistingof 50% wheat protein hydrolysate, 25% free leucineand 25% free phenylalanine was most effective (vanLoon et al. 2000b). Although the effects of carbohy-drate combined with protein on glycogen resyn-thesis after resistance exercise are unknown, there does appear to be a beneficial effect of ingestingsome protein and/or amino acids in combinationwith carbohydrate on glycogen resynthesis aftersubmaximal cycling exercise compared to the sameamount of carbohydrate only (Zawadzki et al. 1992;van Loon et al. 2000a). This effect is likely due togreater insulin secretion after combined carbo-hydrate and protein intake although it is pointedout that when carbohydrate intake is very high

(1.2 g·kg–1·h–1), additional protein does not furtherenhance the rate of glycogen resynthesis ( Jentjens et al. 2001). Enhanced insulin levels resulting fromcarbohydrate combined with protein could beexpected to have a favorable effect on net proteinbalance because insulin is generally accepted as astimulator of protein synthesis when adequateamino acids are available (Kimball & Jefferson 2002;Kimball et al. 2002).

Implications of diet-induced hormonalchanges on body composition

A goal of many athletes and individuals in generalis to improve body composition, that is, to decreaseadipose tissue (fat mass) and/or increase skeletalmuscle tissue (lean body mass). Hormones aremajor regulators of protein turnover in skeletalmuscle and triacylglycerol turnover in adipose tissue. As discussed above, quantity, quality andtiming of dietary intake modulates many of the hormones that regulate protein and triacylglycerolturnover. Thus, from a theoretical perspective feed-ing can be viewed as a strategy to alter the hormonalenvironment to alter protein and lipid balance,which over time could lead to decreased fat massand/or increased lean body mass.

Predicting the impact of a defined meal on proteinand lipid balance and eventually body compositionis however quite complex. Consider that most hor-mones, at least those discussed in this article, affectboth protein and lipid balance through multiplemechanisms (Fig. 29.2), and that unlike the situationin vitro, hormones change in vivo simultaneously inresponse to feeding. Thus, the interactions amongvarious hormones determine the overall response inboth skeletal muscle and adipose tissue. Because ofthese multiple interactions, translation of the infor-mation known about postprandial and exercise-induced hormonal responses into specific dietaryrecommendations aimed at improving body com-position is a complicated task. Nutritional strategiesduring the time periods before and after exercisehave been particularly studied and shown to haverelatively predictable affects on hormones and pro-tein and lipid metabolism (Volek 2004), so this is an appropriate starting point for discussing the

Page 452: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

436 chapter 29

implications of diet-induced hormonal responses.Theoretically, increases in GH, IGF-I and testoster-one would all have favorable effects on skeletalmuscle and adipose tissue balance; that is, elevationsin these hormones would contribute to increasedlean body mass and decreased fat mass. Elevatedcortisol would be negative in terms of skeletal muscle but might contribute to enhanced lipolysis.In contrast, elevated insulin would have positiveeffects on skeletal muscle and negative effects onlipid balance.

Nutrients, hormones, and skeletal muscle protein balance

Exercise, particularly resistance training, stimulates

protein synthesis and protein breakdown, the bal-ance of which determines the anabolic response ofmuscle to resistance exercise. Quantity, quality andtiming of dietary intake after exercise influencesnutrient and hormone availability at specific recep-tors at target tissues (i.e. skeletal muscle and adip-ose tissue). The contraction-induced mechanicaland chemical events in muscle interact with nutrientand hormonal signals to regulate enzymes (e.g.glycogen synthase) and mediate gene level tran-scription and translation of proteins (Turner et al.1988). Exercise also results in increased blood flowto the active skeletal muscles, which has importantimplications for pre-exercise meals that couldenhance hormone interactions and the delivery ofnutrients to target receptors during and after exer-

lipolysislipogenesis

lipogenesis

LPL expression

LPL expression

LPL expression

LPL expression

amino acid oxidationPRO synthesis

PRO breakdown

PRO breakdown

amino acid uptake

PRO breakdown

Adiposetissue

balance

FATAG

SkeletalmusclebalanceGH

Lipogenesis

Lipolysis

glucose transport/oxidation

glucose uptake/oxidation

lipolysis

lipolysis

PRO synthesisPRO breakdown?

IGF-I

(–)

(+)

(+)

(+)

PRO synthesisPRO breakdown?

Testosterone(–) (+)

lipolysis

Cortisol(–) (–)

PRO synthesis

Insulin(+) (+)

AAPRO

PRO synthesis

PRO breakdown

Fig. 29.2 Hormonal regulation of body composition. Several hormones including growth hormone (GH), insulin-likegrowth factor I (IGF-I) and its binding proteins, testosterone, cortisol and insulin regulate both skeletal muscle protein(PRO) and adipose tissue balance through multiple mechanisms. Feeding affects the postprandial and exercise-inducedconcentrations of these hormones, which regulate PRO and amino acid (AA) cycling (i.e. protein synthesis/breakdown) in skeletal muscle and triacylglycerol (TAG) and fatty acid (FA) cycling (i.e. lipogenesis/lipolysis) in adipose tissue. Thesehormones do not change in isolation, rather the simultaneous interaction among hormones must be considered in terms oftheir ultimate impact on protein and lipid balance and lean body mass and fat mass.

Page 453: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 437

cise. The combined effect of muscular contractionand the increased availability of hormones andnutrients have the potential to enhance the rate ofamino acid and glucose uptake, and promote an anabolic environment. Nutrient availability iscritical during this time as evidenced by studiesshowing little glycogen resynthesis (Pascoe et al.1993; Roy & Tarnopolsky 1998) and a negative pro-tein balance (Biolo et al. 1995, 1997) in the absence ofnutritional intake after exercise. An anabolic nutri-tional and hormonal milieu favorably affects thebalance of protein synthesis/degradation, whichsets the stage for greater protein accretion and muscle fiber hypertrophy with chronic resistancetraining. The resulting increased force productioncapabilities can improve the intensity of subsequentworkouts and further enhance the resistance exer-cise stimuli in a feedforward fashion. Eventually,the repeated exposures to resistance exercise work-outs will lead to measurable increases in musclestrength and size.

Feeding has been shown to be a simple and effect-ive method to alter rates of protein synthesis(Svanberg et al. 2000). Infusion of amino acids orexogenous administration of amino acids with or without carbohydrate stimulates protein syn-thesis after exercise (Bennet et al. 1989; Biolo et al.1997; Tipton et al. 1999a, 1999b; Rasmussen et al.2000). Carbohydrate intake after resistance exercisedecreases measures of protein breakdown andslightly increases fractional muscle protein syn-thetic rate, which is likely due primarily to increasesin insulin (Roy et al. 1997). Protein synthesis wasstimulated ∼ 400% above pre-exercise values whena protein and carbohydrate supplement (6 g essen-tial amino acids and 35 g sucrose) was consumed 1 or 3 h after resistance exercise (Rasmussen et al.2000). Consumption of this same protein and car-bohydrate supplement immediately before exerciseresulted in increased amino acid delivery to muscleand even greater net muscle protein synthesis(Tipton et al. 2001). Essential amino acids have beenshown to be primary regulators of muscle proteinsynthesis with little contribution from non-essentialamino acids (Smith, K. et al. 1998; Tipton et al. 1999a,1999b). The branched-chain amino acids, particu-larly leucine, appear to be the most important stimu-

lators of skeletal muscle protein synthesis (Kimball& Jefferson 2002). Recent work indicates that it is the extracellular levels of essential amino acids inthe blood that regulate muscle protein synthesis as opposed to intramuscular amino acids (Bohe et al. 2003). Whether these acute changes in skeletalmuscle protein metabolism reflect chronic changesin lean body mass is unknown.

A recent study in elderly men investigated theeffect of timing of protein and carbohydrate supple-mentation on muscle size and strength responses to12 week of resistance training (Esmarck et al. 2001).The supplement (10 g protein, 7 g carbohydrate)was consumed immediately or 2 h after each train-ing session. The group who ingested the supple-ment immediately after exercise had significantlygreater increases in lean body mass, muscle fiberarea and quadriceps femoris area. These data indic-ate that altering the timing of calories, which affectsthe nutrient and hormonal milieu, can impactchronic adaptation to training. Specifically, earlyintake of protein and carbohydrate after a workoutis more effective at increasing skeletal musclehypertrophy and lean body mass than a supplementconsumed later. These findings are in conflict with a study that showed no differences in acute meas-ures of protein balance when protein was ingested 1 or 3 h after exercise in healthy young subjects(Rasmussen et al. 2000). This apparent discrepancyrelated to timing of protein ingestion highlights theimportance of linking acute studies that measureprotein kinetics to long-term training studies thatassess outcome measures related to muscle size. The only study to date that has done this is workshowing that carbohydrate intake during resistanceexercise was an effective strategy to lower cortisollevels and that the magnitude of cortisol reductionwas directly related to measures of muscle fiberhypertrophy (Tarpenning et al. 2001).

Practically no work has examined how acute diet-induced changes in testosterone, GH or IGF-Imight contribute to the acute changes in protein bal-ance and chronic changes in lean body mass. Ourlaboratory has discovered that circulating testoster-one is consistently modulated by nutrient intakeand that a mixed meal alters post-exercise skeletalmuscle androgen content, indicating a possible link

Page 454: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

438 chapter 29

between diet, testosterone and skeletal muscle pro-tein metabolism.

In summary, there appears to be an interactionbetween increased availability of amino acids, in-creased insulin and possibly other hormones afterexercise and the timing of supplement ingestion (i.e.immediately before exercise) may be important tomaximize the anabolic response (Esmarck et al.2001; Tipton et al. 2001). Consumption of a proteinand carbohydrate supplement at times around exer-cise (i.e. immediately before and immediately afterexercise) may provide the ideal anabolic situationfor muscle growth. There does not appear to be anyvalue in including fat in the pre- or post-exercisemeals from a hormonal or metabolic perspective.

Nutrients, hormones and adipose tissuetriacylglycerol balance

Many of the studies designed to examine how feed-ing affects hormonal responses to exercise havefocused on enhancing protein balance or stimulat-ing glycogen formation with little attention directedto the impact on adipose tissue lipid metabolism.High-carbohydrate intakes are often encouraged tostimulate glycogen synthase and glycogen forma-tion and even protein balance after exercise. Thehormonal environment after carbohydrate feeding,dominated by a surge in insulin, would inhibit lipo-lysis and potentially stimulate lipogenesis in adiposetissue, a scenario not desirable if fat loss is a majorgoal. A better alternative would be to focus on highquality protein with all the essential amino acidsand perhaps low-glycemic carbohydrates duringthe post-exercise period to stimulate protein synthe-sis while keeping insulin low to prevent inhibitionof lipolysis. Protein intake after exercise may have aslight effect on enhancing GH, which would furthersupport lipolysis and decrease lipogenesis. Sub-sequent meals throughout the day should focus onfoods with a low-to-moderate glycemic load to keepinsulin levels low. Since the addition of protein andfat to a meal slow the pace of digestion and lowerthe glycemic load, including quality sources of pro-tein and healthy unsaturated fat should also be apriority.

The popularity of diets that restrict carbohydrate

have increased dramatically in recent years in partbecause they are advertised to be more effective inpromoting weight and fat loss. There were severalstudies performed in the 1960s and 1970s thatshowed greater weight loss with a very low-carbohydrate compared to a low-fat diet, even whendiets contained the same energy content (Rabast et al. 1979), suggesting a metabolic advantage (i.e. agreater weight loss if carbohydrates are low com-pared to isoenergetic diets of different macronutri-ent composition). Very little follow-up work wasdone until recently, as evidenced by several publica-tions in 2003 again showing greater weight loss withvery low-carbohydrate diets 3–6 months in dura-tion (Brehm et al. 2003; Foster et al. 2003; Samaha et al. 2003; Sondike et al. 2003). Some early reportsshow that very low-carbohydrate diets result inpreferential loss of fat and preservation of lean bodymass (Benoit et al. 1965; Young et al. 1971; Willi et al.1998; Meckling et al. 2002), suggestive of a nutrientpartitioning effect. In accordance with this notion,we recently reported that a free-living 6-week very low-carbohydrate diet resulted in significantdecreases in fat mass and increases in lean bodymass in normal-weight men (Volek et al. 2002). In afollow-up study we showed that a very low-carbo-hydrate diet resulted in twofold greater whole bodyfat loss and threefold greater fat loss in the trunkregion compared to a low-fat diet (Volek et al. 2004).Carbohydrate restricted diets remain controversial(Blackburn et al. 2001; Freedman et al. 2001), yet evidence indicates they are very effective in theshort-term and are not associated with any adverseeffects (Volek et al. 2000, 2003; Sharman et al. 2002;Volek & Westman 2002).

Very low-carbohydrate diets are low glycemicload diets. Although the mechanisms by which verylow-carbohydrate diets benefit weight and fat losshas not elucidated, a reduction in insulin is probablyimportant in explaining a portion of the greater fatloss (Volek et al. 2002). Inhibition of lipolysis occursat relatively low concentrations of insulin with ahalf-maximal effect occurring at a concentration of12 pmol·L–1 and a maximal effect at a concentrationof about 200–300 pmol·L–1 (Jensen et al. 1989). Thus,even small reductions in insulin may be permissiveto mobilization of body fat on a low glycemic diet. A

Page 455: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 439

metabolic advantage, possibly driven by increasedprotein turnover to supply alanine for gluconeogen-esis, is another plausible hypothesis to explaingreater weight loss on very low-carbohydrate diets(Feinman & Fine 2004). Although the benefits of following a low glycemic load diet or a low-carbohydrate, moderate protein and fat diet on fatloss look promising, the ideal diet for fat loss andgeneral health remains controversial. However,overwhelming evidence indicates controlling insulinlevels through diet is important.

Summary

It is clear that feeding and meal composition sig-nificantly alter postprandial and post-exercise hor-monal responses. Yet the importance of acute andchronic effects of these hormonal changes on skel-etal muscle and adipose tissue balance and, ulti-mately, body composition remain largely unknown.

Given the important regulatory functions of hor-mones on muscle and lipid balance, the influence ofdiet on endocrine function is arguably of consider-able importance. Optimizing the hormonal environ-ment in favor of an anabolic profile during therecovery period between exercise sessions would beadvantageous for individuals attempting to max-imize gains in muscle size and strength. Specific-ally, by elevating the primary anabolic hormonesinvolved in muscle tissue growth (i.e. testosterone,GH, insulin and IGF-I) and/or decreasing majorcatabolic hormones (i.e. cortisol) a hormonal milieumaximizing protein balance and muscle hyper-trophy would be created. If fat loss is a goal, then itwould be advantageous to keep insulin levels lowand GH and testosterone levels elevated. From ahormonal perspective, meals with a low glycemicload with moderate amounts of quality protein andhealthy unsaturated fat would be conducive for fatloss and general health.

ReferencesAizawa, H. & Niimura, M. (1996) Mild

insulin resistance during oral glucosetolerance test (OGTT) in women withacne. Journal of Dermatology 23, 526–529.

Al-Damluji, S., Iveson, T., Thomas, J.M. et al. (1987) Food-induced cortisolsecretion is mediated by central α1adrenoceptor modulation of pituitaryACTH secretion. Clinical Endocrinology26, 629–636.

Anthony, T.G., Anthony, J.C., Lewitt, M.S.,Donovan, S.M. & Layman, D.K. (2001)Time course changes in IGFBP-1 aftertreadmill exercise and postexercise food intake in rats. American Journal ofPhysiology. Endocrinology and Metabolism280, E650–E656.

Bacurau, R.F., Bassit, R.A., Sawada, L. et al.(2002) Carbohydrate supplementationduring intense exercise and the immuneresponse of cyclists. Clinical Nutrition(Edinburgh, Lothian) 21, 423–429.

Baker, H.W., Best, J.B., Burger, H.G. &Cameron, D.P. (1972) Plasma humangrowth hormone levels in response tomeals: a reappraisal. Australian Journal ofExperimental Biology and Medical Science50, 715–724.

Bennet, W.M., Connacher, A.A.,Scrimgeor, C.M., Smith, K. & Bennie,M.J. (1989) Increase in anterior tibialismuscle protein synthesis in healthy man

during mixed amino acid infusion:studies of incorporation of [1-13C]leucine. Clinical Science 76, 447–454.

Benoit, F.L., Martin, R.L. & Watten, R.H.(1965) Changes in body compositionduring weight reduction in obesity.Annals of Internal Medicine 63, 604–612.

Bereket, A., Wilson, T.A., Blethen, S.L. et al.(1996) Effect of short-term fasting onfree/dissociable insulin-like growthfactor I concentrations in normal humanserum. Journal of Clinical Endocrinologyand Metabolism 81, 4379–4384.

Bergh, C., Carlsson, B., Olsson, J.H.,Selleskog, U. & Hillensjo, T. (1993)Regulation of androgen production incultured human thecal cells by insulin-like growth factor I and insulin. Fertilityand Sterility 59, 323–331.

Bernardi, F., Petraglia, F., Seppala, M. et al. (1999) GH, IGFBP-1, and IGFBP-3response to oral glucose tolerance test inperimenopausal women: no influence ofbody mass index. Maturitas 33, 163–169.

Biolo, G., Maggi, S.P., Williams, B.D.,Tipton, K.D. & Wolfe, R.R. (1995)Increased rates of muscle proteinturnover and amino acid transport afterresistance exercise in humans. AmericanJournal of Physiology 268, E514–E520.

Biolo, G., Tipton, K.D., Klein, S. & Wolfe,R.R. (1997) An abundant supply of

amino acids enhances the metaboliceffect of exercise on muscle protein.American Journal of Physiology.Endocrinology and Metabolism 273,E122–E129.

Bishop, N.C., Blannin, A.K., Robson, P.J.,Walsh, N.P. & Gleeson, M. (1999) Theeffects of carbohydrate supplementationon immune responses to a soccer-specific exercise protocol. Journal ofSports Sciences 17, 787–796.

Bishop, N.C., Blannin, A.K., Walsh, N.P. & Gleeson, M. (2001) Carbohydratebeverage ingestion and neutrophildegranulation responses followingcycling to fatigue at 75% Vo2max.International Journal of Sports Medicine22, 226–231.

Bishop, N.C., Gleeson, M., Nicholas, C.W.& Ali, A. (2002) Influence ofcarbohydrate supplementation onplasma cytokine and neutrophildegranulation responses to highintensity intermittent exercise.International Journal of Sport Nutrition andExercise Metabolism 12, 145–156.

Blackard, W.G., Hull, E.W. & Lopez, A.(1971) Effect of lipids on growthhormone secretion in humans. Journal of Clinical Investigation 50, 1439–1443.

Blackburn, G.L., Phillips, J.C. & Morreale,S. (2001) Physician’s guide to popular

Page 456: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

440 chapter 29

low-carbohydrate weight-loss diets.Cleveland Clinic Journal of Medicine 68,761–774.

Bloomer, R.J., Sforzo, G.A. & Keller, B.A.(2000) Effects of meal form andcomposition on plasma testosterone,cortisol, and insulin following resistanceexercise. International Journal of SportNutrition and Exercise Metabolism 10,415–424.

Bohe, J., Low, A., Wolfe, R.R. & Rennie,M.J. (2003) Human muscle proteinsynthesis is modulated by extracellularnot intracellular amino acid availability:a dose response study. Journal ofPhysiology 552(Pt. 1), 315–324.

Bonen, A., Belcastro, A.N., MacIntyre, K. &Gardner, J. (1980) Hormonal responsesduring intense exercise preceded byglucose ingestion. Canadian Journal ofApplied Sport Sciences 5, 85–90.

Brand-Miller, J.C., Thomas, M., Swan, V. et al. (2003) Physiological validation ofthe concept of glycemic load in leanyoung adults. Journal of Nutrition 133,2728–2732.

Brehm, B.J., Seely, R.J., Daniels, S.R. &D’Alessio, D.A. (2003) A randomizedtrial comparing a very low-carbohydratediet and a calorie-restricted low-fat dieton body weight and cardiovascular riskfactors in healthy women. Journal ofClinical Endocrinology and Metabolism 88,1617–1623.

Cappon, J.P., Ipp, E., Brasel, J.A. & Cooper,D.M. (1993) Acute effects of high fat and high glucose meals on the growthhormone response to exercise. Journal ofClinical Endocrinology and Metabolism 76,1418–1422.

Cappon, J.P., Brasel, J.A., Mohan, S. &Cooper, D.M. (1994) Effect of briefexercise on circulating insulin-likegrowth factor I. Journal of AppliedPhysiology 76, 2490–2496.

Cara, J.F. & Rosenfield, R.L. (1988) Insulin-like growth factor I and insulinpotentiate luteinizing hormone-inducedandrogen synthesis by rat ovarianthecal-interstitial cells. Endocrinology123, 733–739.

Carli, G., Bonifazi, M., Lodi, L. et al. (1992)Changes in the exercise-inducedhormone response to branched chainamino acid administration. EuropeanJournal of Applied Physiology andOccupational Physiology 64, 272–277.

Chandler, R.M., Byrne, H.K., Patterson,J.G. & Ivy, J.L. (1994) Dietarysupplements affect the anabolichormones after weight-training exercise.Journal of Applied Physiology 76, 839–845.

K.H. (1983) The meal-related peak inplasma cortisol is not mediated byradioimmunoassayable ACTH. Clinical Physiology and Biochemistry 1,329–333.

Feinman, R.D. & Fine, E.J. (2004) ‘A calorieis a calorie’ violates the second law ofthermodynamics. Nutrition Journal 28, 9.

Fineberg, E.S., Horland, A.A. & Merimee,T.J. (1972) Free fatty acid concentrationsand growth hormone secretion in man.Metabolism 21, 491–498.

Follenius, M., Brandenberger, G. & Hietter,B. (1982) Diurnal cortisol peaks and theirrelationships to meals. Journal of ClinicalEndocrinology and Metabolism 55,757–761.

Foster, G.D., Wyatt, H.R., Hill, J.O. et al.(2003) A randomized trial of a low-carbohydrate diet for obesity. NewEngland Journal of Medicine 348,2082–2090.

Foster-Powell, K., Holt, S.H. & Brand-Miller, J.C. (2002) International table of glycemic index and glycemic loadvalues, 2002. American Journal of ClinicalNutrition 76, 5–56.

Freedman, M.R. (2001) Popular diets: ascientific review. Obesity Research 1,1S–40S.

Frystyk, J., Grofte, T., Skjaerbaek, C. &Orskov, H. (1997) The effect of oralglucose on serum free insulin-likegrowth factor-I and -II in health adults.Journal of Clinical Endocrinology andMetabolism 82, 3124–3127.

Frystyk, J., Nyholm, B., Skjaerbaek, C. et al. (2003) The circulating IGF systemand its relationship with 24-h glucoseregulation and insulin sensitivity inhealthy subjects. Clinical Endocrinology58, 777–784.

Galbo, H., Holst, J.J. & Christensen, N.J.(1979) The effects of different diets andof insulin on the hormonal response toprolonged exercise. Acta PhysiologicaScandinavica 107, 19–32.

Gibson, E.L., Checkley, S., Papadopoulos,A. et al. (1999) Increased salivary cortisolreliably induced by a protein-richmidday meal. Psychosomatic Medicine 61,214–224.

Gonzalez-Bono, E., Rohleder, N.,Hellhammer, D.H., Salvador, A. &Kirschbaum, C. (2002) Glucose but notprotein or fat load amplifies the cortisolresponse to psychosocial stress.Hormones and Behavior 41, 328–333.

Green, K.J., Croaker, S.J. & Rowbottom,D.G. (2003) Carbohydratesupplementation and exercise-inducedchanges in T-lymphocyte function.

Chromiak, J.A. & Antonio, J. (2002) Use of amino acids as growth hormone-releasing agents by athletes. Nutrition 18,657–661.

De Pergola, G. (2000) The adipose tissuemetabolism: role of testosterone anddehydroepiandrosterone. InternationalJournal of Obesity and Related MetabolicDisorders 24, S59–S63.

Deuster, P.A., Singh, A., Hofmann, A.,Moses, F.M. & Chrousos, G.C. (1992)Hormonal responses to ingesting wateror a carbohydrate beverage during a 2 hrun. Medicine and Science in Sports andExercise 24, 72–79.

Diamond, M.P., Grainger, D.A., Laudano,A.J., Starick-Zych, K. & DeFronzo, R.A.(1991) Effect of acute physiologicalelevations of insulin on circulatingandrogen levels in nonobese women.Journal of Clinical Endocrinology andMetabolism 72, 883–887.

Divertie, G.D., Jensen, M.D. & Miles, J.M.(1991) Stimulation of lipolysis in humans by physiologicalhypercortisolemia. Diabetes 40,1228–1232.

Djurhuus, C.B., Gravholt, C.H., Nielsen, S.et al. (2002) Effects of cortisol on lipolysisand regional interstitial glycerol levels inhumans. American Journal of Physiology.Endocrinology and Metabolism 283,E172–E177.

Ebeling, P., Stenman, U.H., Seppala, M. & Koivisto, V.A. (1995) Acutehyperinsulinemia, androgenhomeostasis and insulin sensitivity inhealthy man. Journal of Endocrinology146, 63–69.

Ebenbichler, C.F., Kirchmair, R., Egger, C.& Patsch, J.R. (1995) Postprandial stateand atherosclerosis. Current Opinion inLipidology 6, 286–290.

Esmarck, B., Andersen, J.L., Olsen, S. et al. (2001) Timing of post-exerciseprotein intake is important for musclehypertrophy with resistance training inelderly humans. Journal of Physiology 535,301–311.

Etherton, T.D. (2000) The biology ofsomatotropin in adipose tissue growthand nutrient partitioning. Journal ofNutrition 130, 2623–2625.

Falcone, T., Finegood, D.T., Fantus, I.G. &Morris, D. (1990) Androgen response to endogenous insulin secretion duringthe frequently sampled intravenousglucose tolerance test in normal andhyperandrogenic women. Journal ofClinical Endocrinology and Metabolism 71,1653–1657.

Fehm, H.L., Holl, R., Klein, E. & Voigt,

Page 457: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 441

Journal of Applied Physiology 95,1216–1223.

Habito, R.C. & Ball, M.J. (2001)Postprandial changes in sex hormonesafter meals of different composition.Metabolism 50, 505–511.

Haffner, S.M., Valdez, R.A., Mykkanen, L.,Stern, M.P. & Katz, M.S. (1994)Decreased testosterone anddehydroepiandrosterone sulfateconcentrations are associated withincreased insulin and glucoseconcentrations in nondiabetic men.Metabolism 43, 599–603.

Heffernan, M.A., Jiang, W.J., Thorburn,A.W. & Ng, F.M. (2000) Effects of oraladministration of a synthetic fragmentof human growth hormone on lipidmetabolism. American Journal ofPhysiology. Endocrinology and Metabolism279, E501–E507.

Henson, D.A., Nieman, D.C., Parker, J.C. et al. (1998) Carbohydratesupplementation and the lymphocyteproliferative response to long endurancerunning. International Journal of SportsMedicine 19, 574–580.

Henson, D.A., Nieman, D.C., Nehlsen-Cannarella, S.L. et al. (2000) Influence of carbohydrate on cytokine andphagocytic responses to 2 h of rowing.Medicine and Science in Sports and Exercise32, 1384–1389.

Hjalmarsen, A., Aasebo, U., Aakvaag, A. &Jorde, R. (1996) Sex hormone responsesin healthy men and male patients withchronic obstructive pulmonary diseaseduring an oral glucose load.Scandinavian Journal of Clinical andLaboratory Investigation 56, 635–640.

Holden, J.P., Butzow, T.L., Laughlin, G.A.et al. (1995) Regulation of insulin-likegrowth factor binding protein-1 duringthe 24-hour metabolic clock and inresponse to hypoinsulinemia induced by fasting and Sandostatin in normalwomen. Journal of the Society forGynecologic Investigation 2, 38–44.

Holt, R.I., Simpson, H.L. & Sonksen, P.H.(2003) The role of the growth hormone-insulin-like growth factor axis in glucosehomeostasis. Diabetic Medicine 20, 3–15.

Hopkins, N.J., Jakeman, P.M., Hughes, S.C. & Holly, J.M. (1994) Changes incirculating insulin-like growth factor-binding protein-1 (IGFBP-1) duringprolonged exercise: effect ofcarbohydrate feeding. Journal of ClinicalEndocrinology and Metabolism 79,1887–1890.

Ishizuka, B., Quigley, M.E. & Yen, S.S.(1983) Pituitary hormone release in

International Journal of Sport Nutrition andExercise Metabolism 11, 149–161.

Koistinen, H., Koistinen, R., Selenius, L.,Ylikorkala, Q. & Seppala, M. (1996)Effect of marathon run on serum IGF-Iand IGF-binding protein 1 and 3 levels.Journal of Applied Physiology 80, 760–764.

Kraemer, W.J., Volek, J.S., Bush, J.A.,Putukian, M. & Sebastianelli, W.J. (1998)Hormonal responses to consecutivedays of heavy-resistance exercise with or without nutritional supplementation.Journal of Applied Physiology 85,1544–1555.

Lavoie, J.M., Fillion, Y., Couturier, K. &Corriveau, P. (2002) Evidence that thedecrease in liver glycogen is associatedwith the exercise-induced increase inIGFBP-1. Journal of Applied Physiology 93,798–804.

Leal, A.M. & Moreira, A.C. (1997) Foodand the circadian activity of thehypothalamic–pituitary–adrenal axis.Brazilian Journal of Medical and BiologicalResearch 30, 1391–1405.

Lee, P.D., Conover, C.A. & Powell, D.R.(1993) Regulation and function ofinsulin-like growth factor-bindingprotein-1. Proceedings of Society forExperimental Biology and Medicine 204,4–29.

Lee, P.D., Giudice, L.C., Conover, C.A. &Powell, D.R. (1997) Insulin-like growthfactor binding protein-1: recent findingsand new directions. Proceedings of Societyfor Experimental Biology and Medicine 216,319–357.

van Loon, L.J., Saris, W.H., Kruijshoop, M.& Wagenmakers, A.J. (2000a)Maximizing postexercise muscleglycogen synthesis: carbohydratesupplementation and the application of amino acid or protein hydrolysatemixtures. American Journal of ClinicalNutrition 72, 106–111.

van Loon, L.J., Saris, W.H., Verhagen, H. & Wagenmakers, A.J. (2000b) Plasmainsulin responses after ingestion ofdifferent amino acid or protein mixtureswith carbohydrate. American Journal ofClinical Nutrition 72, 96–105.

van Loon, L.J., Kruijshoop, M., Menheere,P.P. et al. (2003) Amino acid ingestionstrongly enhances insulin secretion inpatients with long-term type 2 diabetes.Diabetes Care 26, 625–630.

Meckling, K.A., Gauthier, M., Grubb, R. & Sanford, J. (2002) Effects of ahypocaloric, low-carbohydrate diet on weight loss, blood lipids, bloodpressure, glucose tolerance, and bodycomposition in free-living overweight

response to food ingestion: evidence forneuroendocrine signals from gut tobrain. Journal of Clinical Endocrinologyand Metabolism 57, 1111–1116.

Ivandic, A., Prpic-Krizevac, I., Jakic, M. & Bacun, T. (1999) Changes in sexhormones during an oral glucosetolerance test in healthy premenopausalwomen. Fertility and Sterility 71, 268–273.

Jenkins, D.J., Wolever, T.M., Taylor, R.H. et al. (1981) Glycemic index of foods: aphysiological basis for carbohydrateexchange. American Journal of ClinicalNutrition 34, 362–366.

Jensen, M.D., Caruso, M., Heiling, V.J. &Miles, J.M. (1989) Insulin regulation oflipolysis in nondiabetic and IDDMsubjects. Diabetes 38, 1595–1601.

Jentjens, R.L., van Loon, L.J., Mann, C.H.,Wagenmakers, A.J. & Jeukendrup, A.E.(2001) Addition of protein and aminoacids to carbohydrates does not enhancepostexercise muscle glycogen synthesis.Journal of Applied Physiology 91, 839–846.

Jezova-Repcekova, D., Vigas, M. & Klimes,I. (1980) Decreased plasma cortisolresponse to pharmacological stimuliafter glucose load in man. EndocrinologiaExperimentalis 14, 113–120.

Johannessen, A., Hagen, C. & Galbo, H.(1981) Prolactin, growth hormone,thyrotropin, 3,5,3’-triiodothyronine, andthyroxine responses to exercise after fat-and carbohydrate-enriched diet. Journalof Clinical Endocrinology and Metabolism52, 56–61.

Kersten, S. (2001) Mechanisms ofnutritional and hormonal regulation oflipogenesis. EMBO Reports 2, 282–286.

Kimball, S.R. & Jefferson, L.S. (2002)Control of protein synthesis by aminoacid availability. Current Opinion inClinical Nutrition and Metabolic Care 5,63–67.

Kimball, S.R., Farrell, P.A. & Jefferson, L.S.(2002) Invited review: role of insulin intranslational control of protein synthesisin skeletal muscle by amino acids orexercise. Journal of Applied Physiology 93,1168–1180.

Knoll, E., Muller, F.W., Ratge, D.,Bauersfeld, W. & Wisser, H. (1984)Influence of food intake onconcentrations of plasmacatecholamines and cortisol. Journal of Clinical Chemistry and ClinicalBiochemistry 22, 597–602.

Koch, A.J., Potteiger, J.A., Chan, M.A.,Benedict, S.H. & Frey, B.B. (2001)Minimal influence of carbohydrateingestion on the immune responsefollowing acute resistance exercise.

Page 458: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

442 chapter 29

women. Canadian Journal of Physiologyand Pharmacology 80, 1095–1105.

Meikle, A.W., Benson, S.J., Liu, X.H.,Boam, W.D. & Stringham, J.D. (1989)Nonesterified fatty acids modulatesteroidogenesis in mouse Leydig cells.American Journal of Physiology 257,E937–E942.

Meikle, A.W., Stringham, J.D., Woodward,M.G. & McMurry, M.P. (1990) Effects ofa fat-containing meal on sex hormonesin men. Metabolism 39, 943–946.

Miller, S.L., Maresh, C.M., Armstrong, L.E. et al. (2002) Metabolic response to provision of mixed protein–carbohydrate supplementation duringendurance exercise. International Journalof Sport Nutrition and Exercise Metabolism12, 384–397.

Mitchell, J.B., Costill, D.L., Houmard, J.A.et al. (1990) Influence of carbohydrateingestion on counterregulatoryhormones during prolonged exercise.International Journal of Sports Medicine 11,33–36.

Mitchell, J.B., Pizza, F.X., Paquet, A. et al.(1998) Influence of carbohydrate statuson immune responses before and afterendurance exercise. Journal of AppliedPhysiology 84, 1917–1925.

Murray, R., Paul, G.L., Seifert, J.G. & Eddy,D.E. (1991) Responses to varying rates ofcarbohydrate ingestion during exercise.Medicine and Science in Sports and Exercise23, 713–718.

Murray, R., Bartoli, W.P., Eddy, D.E. &Horn, M.K. (1995) Physiological andperformance responses to nicotinic-acidingestion during exercise. Medicine and Science in Sports and Exercise 27,1057–1062.

Nakagawa, E., Nagaya, N., Okumura, H. et al. (2002) Hyperglycaemia suppressesthe secretion of ghrelin, a novel growth-hormone-releasing peptide: responses tothe intravenous and oral administrationof glucose. Clinical Science (London,England) 103, 325–328.

Nehlsen-Cannarella, S.L., Fagoaga, O.R.,Nieman, D.C. et al. (1997) Carbohydrateand the cytokine response to 2.5 h ofrunning. Journal of Applied Physiology 82,1662–1667.

Nieman, D.C. (1998) Influence ofcarbohydrate on the immune responseto intensive, prolonged exercise. ExerciseImmunology Review 4, 64–76.

Nieman, D.C., Nehlsen-Cannarella, S.L.,Fagoaga, O.R. et al. (1998) Influence ofmode and carbohydrate on the cytokineresponse to heavy exertion. Medicine and Science in Sports and Exercise 30,671–678.

Nieman, D.C., Davis, J.M., Henson, D.A.

Ricart, W. & Fernandez-Real, J.M. (2001)No decrease in free IGF-I with increasinginsulin in obesity-related insulinresistance. Obesity Research 9, 631–636.

Rojdmark, S., Rydvald, Y., Aquilonius, A.& Brismar, K. (2000) Insulin-like growthfactor (IGF)-1 and IGF-binding protein-1concentrations in serum of normalsubjects after alcohol ingestion: evidencefor decreased IGF-1 bioavailability.Clinical Endocrinology 52, 313–318.

Rooyackers, O.E. & Nair, K.S. (1997)Hormonal regulation of human muscleprotein metabolism. Annual Review ofNutrition 17, 457–485.

Roth, J., Glick, S.M., Yalow, R.S. & Berson,S.A. (1963) Hypoglycemia: a potentstimulus to secretion of growthhormone. Science 140, 987–988.

Roy, B.D. & Tarnopolsky, M.A. (1998)Influence of different macronutrientintakes on muscle glycogen resynthesisafter resistance exercise. Journal ofApplied Physiology 72, 1854–1859.

Roy, B.D., Tarnopolsky, M.A.,MacDougall, J.D., Fowles, J. &Yarasheski, K.E. (1997) Effect of glucose supplement timing on proteinmetabolism after resistance training.Journal of Applied Physiology 82,1882–1888.

Salmeron, J., Manson, J.E., Stampfer, M.J. et al. (1997) Dietary fiber, glycemic load,and risk of non-insulin-dependentdiabetes mellitus in women. JAMA 277,472–477.

Samaha, F.F., Iqbal, N., Seshadri, P. et al.(2003) A low-carbohydrate as comparedwith a low-fat diet in severe obesity. New England Journal of Medicine 348,2074–2081.

Schaller, G., Schmidt, A., Pleiner, J. et al.(2003) Plasma ghrelin concentrations are not regulated by glucose or insulin: a double-blind, placebo-controlledcrossover clamp study. Diabetes 52,16–20.

Sebokova, E., Garg, M.L. & Clandinin,M.T. (1988) Modulation of receptor-mediated gonadotropin action in rattestes by dietary fat. American Journal ofPhysiology 254, E708–E712.

Sebokova, E., Garg, M.L., Wierzbicki, A.,Thompson, A.B.R. & Clandinin, M.T.(1990) Alteration of the lipidcomposition of rat testicular plasmamembranes by dietary (n-3) fatty acidschanges the responsiveness of Leydigcells and testosterone synthesis. Journalof Nutrition 120, 610–618.

Sharman, M.J., Kraemer, W.J., Love, D.M.et al. (2002) A ketogenic diet favorablyaffects serum biomarkers for

et al. (2003) Carbohydrate ingestioninfluences skeletal muscle cytokinemRNA and plasma cytokine levels aftera 3-h run. Journal of Applied Physiology 94,1917–1925.

Nuttall, F.Q., Mooradian, A.D., Gannon,M.C., Billington, C. & Krezowski, P.(1984) Effect of protein ingestion on the glucose and insulin response to astandardized oral glucose load. DiabetesCare 7, 465–470.

Nuttall, F.Q., Gannon, M.C., Wald, J.L. & Ahmed, M. (1985) Plasma glucose and insulin profiles in normal subjectsingesting diets of varying carbohydrate,fat, and protein content. Journal of theAmerican College of Nutrition 4, 437–450.

Nyomba, B.L., Berard, L. & Murphy, L.J.(1997) Free insulin-like growth factor I(IGF-I) in healthy subjects: relationshipwith IGF-binding proteins and insulin sensitivity. Journal of ClinicalEndocrinology and Metabolism 82,2177–2181.

Pascoe, D.D., Costill, D.L., Fink, W.J.,Robergs, R.A. & Zachwieja, J.J. (1993)Glycogen resynthesis in skeletal musclefollowing resistive exercise. Medicine and Science in Sports and Exercise 25,349–354.

Pasquali, R., Macor, C., Vicennati, V. et al.(1997) Effects of acute hyperinsulinemiaon testosterone serum concentrations inadult obese and normal-weight men.Metabolism 46, 526–529.

Patsch, J.R., Miesenbock, G., Hopferwieser,T. et al. (1992) Relation of triglyceridemetabolism and coronary artery disease:studies in the postprandial state.Thrombosis 12, 1336–1345.

Rabast, U., Schonborn, J. & Kasper, H.(1979) Dietetic treatment of obesity witha low and high-carbohydrate diets:comparative studies and clinical results.International Journal of Obesity 3, 201–211.

Rasmussen, B.B., Tipton, K.D., Miller, S.L.,Wolf, S.E. & Wolfe, R.R. (2000) An oralamino acid-carbohydrate supplementenhances muscle protein anabolism afterresistance exercise. Journal of AppliedPhysiology 88, 386–392.

Reynolds, R.M., Walker, B.R., Syddall, H.E.et al. (2001) Elevated plasma cortisol inglucose-intolerant men: differences inresponses to glucose and habituation to venepuncture. Journal of ClinicalEndocrinology and Metabolism 86,1149–1153.

Reynolds, R.M., Syddall, H.E., Walker,B.R., Wood, P.J. & Phillips, D.I. (2003)Predicting cardiovascular risk factorsfrom plasma cortisol measured duringoral glucose tolerance tests. Metabolism52, 524–527.

Page 459: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

impact of diet on hormones 443

cardiovascular disease in normal-weightmen. Journal of Nutrition 132, 1879–1885.

Siddals, K.W., Westwood, M., Gibson, J.M.& White, A. (2002) IGF-binding protein-1 inhibits IGF effects on adipocytefunction: implications for insulin-likeactions at the adipocyte. Journal ofEndocrinology 174, 289–297.

Slag, M.F., Ahmad, M., Gannon, M.C. &Nuttall, F.Q. (1981) Meal stimulation of cortisol secretion: a protein inducedeffect. Metabolism 30, 1104–1108.

Smith, K., Reynolds, N., Downie, S., Patel, A. & Rennie, M.J. (1998) Effects offlooding amino acids on incorporationof labeled amino acids into humanmuscle protein. American Journal ofPhysiology. Endocrinology and Metabolism275, E73–E78.

Smith, S., Ravnikar, V.A. & Barbieri, R.L.(1987) Androgen and insulin response to an oral glucose challenge inhyperandrogenic women. Fertility andSterility 48, 72–77.

Sondike, S.B., Copperman, N. & Jacobson,M.S. (2003) Effects of a low-carbohydratediet on weight loss and cardiovascularrisk factors in overweight adolescents.Journal of Pediatrics 142, 253–258.

Starkie, R.L., Rolland, J., Angus, D.J.,Anderson, M.J. & Febbraio, M.A. (2001)Circulating monocytes are not thesource of elevations in plasma IL-6 andTNF-α levels after prolonged running.American Journal of Physiology. CellPhysiology 280, C769–C774.

Svanberg, E., Ennion, S., Isgaard, J. &Goldspink, G. (2000) Postprandialresynthesis of myofibrillar proteins is translationally rather thantranscriptionally regulated in humanskeletal muscle. Nutrition 16, 42–46.

Tarpenning, K.M., Wiswell, R.A.,Hawkins, S.A. & Marcell, T.J. (2001)Influence of weight training exercise and modification of hormonal responseon skeletal muscle growth. Journal ofScience and Medicine in Sport 4, 431–446.

Tiitinen, A., Pekonen, F., Stenman, U.H. &Laatikainen, T. (1990) Plasma androgensand oestradiol during oral glucosetolerance test in patients with polycysticovaries. Human Reproduction (Oxford,England) 5, 242–245.

Tipton, K.D., Ferrando, A.A., Phillips,S.M., Doyle, D., Jr. & Wolfe, R.R. (1999a)Postexercise net protein synthesis inhuman muscle from orally administeredamino acids. American Journal ofPhysiology 276, E628–E634.

Tipton, K.D., Gurkin, B.E., Martin, S. &Wolfe, R.R. (1999b) Nonessential aminoacids are not necessary to stimulate netmuscle protein synthesis in healthy

et al. (2001) Effects of a high-fat diet on postabsorptive and postprandialtestosterone responses to a fat-rich meal. Metabolism 50, 1351–1355.

Volek, J.S., Sharman, M.J., Love, D.M. et al.(2002) Body composition and hormonalresponses to a carbohydrate-restricteddiet. Metabolism 51, 864–870.

Volek, J.S., Sharman, M.J., Gómez, A.L.,Scheett, T.P. & Kraemer, W.J. (2003) An isoenergetic very low-carbohydratediet is associated with improved serumhigh-density lipoprotein cholesterol(HDL-C), total cholesterol to HDL-Cratio, triacylglycerols, and postprandiallipemic responses compared to a low-fatdiet in normal weight, normolipidemicwomen. Journal of Nutrition 133,2756–2761.

Volek, J.S., Sharman, M.J., Gomez, A.L. et al. (2004) Comparison of very low-carbohydrate and low-fat diet on fastinglipids, LDL subclasses, insulinresistance, and postprandial lipemicresponses in overweight women. Journalof the American College of Nutrition 23(2),177–184.

Walker, B.R., Soderberg, S., Lindahl, B. &Olsson, T. (2000) Independent effects of obesity and cortisol in predictingcardiovascular risk factors in men andwomen. Journal of Internal Medicine 247,198–204.

Whitley, H.A., Humphreys, S.M.,Campbell, I.T. et al. (1998) Metabolic and performance responses duringendurance exercise after high-fat and high-carbohydrate meals. Journal of Applied Physiology 85, 418–424.

Willi, S.M., Oexmann, M.J., Wright, N.M.,Collop, N.A. & Lyndon, L. (1998) Theeffects of a high-protein, low-fat,ketogenic diet on adolescents withmorbid obesity: body composition,blood chemistries, and sleepabnormalities. Pediatrics 101, 61–67.

Williams, A.G., Ismail, A.N., Sharma, A. & Jones, D.A. (2002) Effects of resistanceexercise volume and nutritionalsupplementation on anabolic andcatabolic hormones. European Journal ofApplied Physiology 86, 315–321.

Young, C.M., Scanlan, S.S., Im, H.S. &Lutwak, L. (1971) Effect on bodycomposition and other parameters inobese young men of carbohydrate levelof reduction diet. American Journal ofClinical Nutrition 24, 290–296.

Zawadzki, K.M., Yaspelkis, B.B., 3rd & Ivy,J.L. (1992) Carbohydrate–proteincomplex increases the rate of muscleglycogen storage after exercise. Journal of Applied Physiology 72, 1854–1859.

volunteers. Journal of NutritionalBiochemistry 10, 89–95.

Tipton, K.D., Rasmussen, B.B., Miller, S.L.et al. (2001) Timing of amino acid–carbohydrate ingestion alters anabolicresponse of muscle to resistanceexercise. American Journal of Physiology281, E197–E206.

Tropeano, G., Lucisano, A., Liberale, I. et al.(1994) Insulin, C-peptide, androgens,and β-endorphin response to oralglucose in patients with polycystic ovary syndrome. Journal of ClinicalEndocrinology and Metabolism 78,305–309.

Tsintzas, O.K., Williams, C., Wilson, W. & Burrin, J. (1996) Influence ofcarbohydrate supplementation early inexercise on endurance running capacity.Medicine and Science in Sports and Exercise28, 1373–1379.

Turner, J.D., Rotwein, P., Novakofski, J. & Bechtel, P.J. (1988) Induction ofmessenger RNA for IGF-I and -II duringgrowth hormone-stimulated musclehypertrophy. American Journal ofPhysiology 255, E513–E517.

Ullrich, I.H., Peters, P.J. & Albrink, M.J.(1985) Effect of low-carbohydrate dietshigh in either fat or protein on thyroidfunction, plasma insulin, glucose, andtriglycerides in healthy young adults.Journal of American College of Nutrition 4,451–459.

Utter, A.C., Kang, J., Nieman, D.C. et al.(1999) Effect of carbohydrate ingestionand hormonal responses on ratings ofperceived exertion during prolongedcycling and running. European Journal ofApplied Physiology and OccupationalPhysiology 80, 92–99.

Vicennati, V., Ceroni, L., Gagliardi, L.,Gambineri, A. & Pasquali, R. (2002)Comment: response of thehypothalamic–pituitary–adrenocorticalaxis to high-protein/fat and high-carbohydrate meals in women withdifferent obesity phenotypes. Journal ofClinical Endocrinology and Metabolism 87,3984–3988.

Volek, J.S. (2004) Nutritional practices forresistance training. Medicine and Sciencein Sports and Exercise 36, 689–696.

Volek, J.S. & Westman, E.C. (2002) Lowcarbohydrate weight-loss diets revisited.Cleveland Clinic Journal of Medicine 69,849–862.

Volek, J.S., Gómez, A.L. & Kraemer, W.J.(2000) Fasting and postprandiallipoprotein responses to a low-carbohydrate diet supplemented with n-3 fatty acids. Journal of the AmericanCollege of Nutrition 19, 383–391.

Volek, J.S., Gómez, A.L., Love, D.M.

Page 460: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

444

Introduction

Upon arrival at high altitude, rapid physiologicadjustments occur to compensate for the reductionin ambient oxygen (O2). Immediate physiologicadjustments, which include an increase in ventila-tion and heart rate, provide a rapid first line ofdefense against the reduced ambient O2 (Stenberg et al. 1966; Easton et al. 1986). Longer term physio-logic adaptations, which include continued hyper-ventilation, increased production of RBCs, changesin circulation, increased water and sodium excre-tion, and shifts in substrate utilization, provide asecond line of defense against the sustained hypoxicenvironment (Young, A.J. & Reeves 2002). The autonomic nervous system plays a critical role inmediating many of the immediate physiologicadjustments to altitude but hormonal alterationsplay a role in fine-tuning many of the body’s longer-term adaptations to altitude.

Although several reviews have examined theneurohumoral response to altitude (Hoyt & Honig1996; Ward et al. 2000; Richalet 2001; Swenson 2001;Mazzeo & Reeves 2003), a clear consensus has notbeen reached regarding the role that hormones playin successfully adapting to altitude. Conflictingresults in the literature are primarily due to differ-ences in study design (field versus chamber studies),time point of measurements during exposure (acuteversus chronic), altitude (low versus high elevation),environmental conditions (normobaric hypoxia ver-sus hypobaric hypoxia), gender, (male versus female),physical fitness levels (trained versus untrained),duration of ascent (rapid versus slow) and illness

status (sick versus non-sick). This review thereforewill focus on summarizing the results of numerousstudies that were conducted under a defined set ofconditions in order to develop a clearer understand-ing of the neurohumoral responses and adaptationsduring rest and exercise at altitude.

The studies included in this review were as fol-lows: (a) both chamber and field studies; (b) studiesemploying hypobaric hypoxia; (c) altitudes rangingfrom 3500 to 5500 m; (d) both men and women; (e)both physically trained and untrained; (f ) both rapid and slow ascent; and (g) if the results of sickand non-sick subjects were separated, only resultsfrom non-sick subjects. Acute altitude exposure wasdefined as < = 3 days and chronic altitude exposurewas defined as 4–21 days of altitude exposure. Thisreview will also focus on human studies unless critical points can only be explained by includinganimal work.

Basic concepts

In order to understand the neurohumoral mech-anisms responsible for adaptation to altitude, it isimperative to first understand the basic conceptsassociated with exposure to altitude. First, baromet-ric pressure (PB) decreases with increasing altitude(Fig. 30.1). Although the percentage of O2 in a givenvolume of air at altitude is the same as at sea level(i.e. 20.93%), the ambient partial pressure of O2 (Po2)decreases (Po2 = PB × % O2). Second, the inspiredoxygen pressure (PIo2) is diminished in direct pro-portion to the reduction in barometric pressure (PIo2 = (PB − 47) × %O2), where the PHo2 is taken to

Chapter 30

Neurohumoral Responses and AdaptationsDuring Rest and Exercise at Altitude

BETH A. BEIDLEMAN, JANET E. STAAB AND ELLEN L. GLICKMAN

Page 461: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 445

be 47 Torr at a body temperature of 37°C. Thus, the O2 pressure gradient at each step in the O2 trans-port cascade is reduced upon exposure to altitude(Fig. 30.2). As a result, less O2 is transferred from theenvironment to the cells as the diffusion of O2 isdirectly dependent on the Po2 gradient, accordingto Fick’s law of diffusion. The result is an impairedability to deliver O2 to the tissues of the body uponexposure to altitude. Numerous short- and long-term physiologic adaptations occur to compensatefor this hypoxemia.

Physiologic adaptations to altitude

Ventilatory

Upon acute exposure to altitude, there is an imme-diate increase in ventilation that is dependent on theseverity of hypoxia (Rahn & Otis 1949; Easton et al.1986; Bisgard & Forster 1996). This increase in venti-lation is primarily the result of hypoxic stimulationof the peripheral chemoreceptors (Weil et al. 1970;Lahiri & Delaney 1975; Fitzgerald & Dehghani1982). Immediately following this increase, ventila-tion declines over the next 10–30 min of altitudeexposure. This response has been termed ‘hypoxicventilatory depression’ and is likely due to build upof inhibitory transmitters such as γ-aminobutyricacid or adenosine in the central nervous system(Smith et al. 2001). Within the next few hours to daysof altitude exposure, there is a progressive increasein ventilation that tends to level off by 7–10 days of altitude exposure (Forster et al. 1975). The mech-anisms involved in ventilatory acclimatization toaltitude are highly debated, but thought to be due to a progressive increase in carotid body sensitivityand central nervous system acid-base changes(Severinghaus et al. 1963; Bisgard & Forster 1996;Smith et al. 2001). The increase in ventilation withinitial and continued altitude exposure mitigatesthe obligatory fall in partial pressure of alveolar O2(PAo2) that follows a reduction in PIo2. In fact, ifcompensatory hyperventilation did not occur, thealveolar gas equation predicts that PAo2 would be10 Torr lower at 4000 m. This increase in PAo2 withhyperventilation facilitates O2 loading in the lungsand provides a rapid and continued first line ofdefense against the reduced ambient Po2.

Hematologic

Erythropoietin (described below) increases uponacute exposure to altitude, which stimulates thehematological cascade (Jelkmann 1992). Reticulo-cytes typically appear within 3–5 days (Grover et al.1998; Reeves et al. 2001) but red blood cell (RBC) volume remains unchanged following 10 days ofaltitude exposure (Sawka et al. 1996). Althoughhemoglobin concentration [Hb] clearly increases in

0 2000 4000

Altitude (m)

6000 8000 10 000

800

700

600

500

400

300

200

Bar

om

etri

c p

ress

ure

(m

mH

g)

Fig. 30.1 The relationship between barometric pressure(PB) and altitude (m).

Inspiredair

Alveolargas

Arterialblood

Mixedvenousblood

Sea level4300 m

160

140

120

100

80

60

40

20

o2

(mm

Hg

)

Fig. 30.2 The oxygen transport cascade at sea level and4300 m. (Modified from Fulco & Cymerman 1988.)

Page 462: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

446 chapter 30

lowlanders sojourning at altitudes for 5–12 days(Maher et al. 1974; Horstman et al. 1980; Wolfel et al.1991, 1998), this increase in [Hb] is most likely due tohemoconcentration (i.e. decrease in plasma volume)rather than erythrocyte volume expansion (Sawka et al. 2000). If the altitude sojourn exceeds 1 month,erythrocyte volume expansion occurs (Reynafarje1957; Pugh 1964). The higher RBC volumes observedin high altitude natives compared to lowlandersalso supports increased RBC production follow-ing an extended period of time at altitude (Weil et al. 1968; Sanchez et al. 1970). The increase in RBCvolume and thus Hb, which is the compoundresponsible for binding O2, ultimately improves O2transport capacity at altitude.

Cardiovascular

Heart rate and cardiac output are increased imme-diately upon ascent to altitude (Stenberg et al. 1966;Vogel & Harris, 1967; Wagner et al. 1986). The abruptincrease in cardiac output is due to an increase inheart rate, since stroke volume remains unchanged(Grover et al. 1986; Wolfel & Levine 2001). Sym-pathetic stimulation and parasympathetic with-drawal (described below) both contribute to thisincrease in heart rate. Normally, sympathetic stimu-lation would also elicit peripheral vasoconstriction.However, acute hypoxia overrides sympathetically-mediated vasoconstriction and induces vasodila-tion in all vascular beds except the lung such thattotal peripheral resistance is decreased (Wolfel &Levine 2001). The specific local vasodilator respons-ible for overriding vasoconstriction is not known,but nitric oxide and epinephrine have been sug-gested as potential candidates (Halliwill 2003).Mixed-venous O2 content also decreases with acutealtitude exposure, which indicates increased peri-pheral O2 extraction (Wolfel et al. 1991, 1998). Thus,increases in both blood flow and peripheral O2extraction upon acute exposure to altitude maintaina tight coupling between metabolic supply anddemand under limited O2 conditions.

With chronic altitude exposure, arterial O2 content(Cao2), which is the product of arterial O2 saturation(Sao2) and [Hb] multiplied by 1.34 mL O2·g [Hb]–1, isincreased due to ventilatory and hematological

adaptations (Wolfel et al. 1991, 1998). Stroke volumedecreases due to a decrease in plasma volume and/or increase in total peripheral resistance while heartrate remains elevated (Grover et al. 1986; Wolfel &Levine 2001; Young, A.J. & Reeves 2002). Cardiacoutput declines following chronic altitude exposurebecause of the decrease in stroke volume (Grover et al. 1986; Wolfel & Levine 2001; Young, A.J. &Reeves 2002). The decline in cardiac output offsetsthe increase in Cao2 such that O2 delivery remainsunchanged following chronic altitude exposure(Grover et al. 1976; Bender et al. 1988). Although adeclining blood flow and cardiac output would notappear beneficial, the prolonged capillary transittime may minimize the alveolar–arterial diffusionlimitation at altitude such that Sao2 is not comprom-ised at the expense of increased blood flow (Boushelet al. 2001).

Fluid regulation

Water and sodium excretion are increased in norm-ally acclimatizing lowlanders with both acute andchronic exposure to altitude (Hoyt & Honig 1996).This natriureis and diuresis elicits a depletion ofplasma volume and increase in [Hb] due to hemo-concentration (Sawka et al. 2000), which counterbal-ances the reduced supply of O2 to the tissues. Thediuresis may also protect against altitude illness(Singh et al. 1969) due to a decrease in extracellularfluid volume and brain edema (Hackett 1999). Themechanisms underlying the natriuretic and diureticeffect of hypoxia are not well understood, but suggestthat hypoxic stimulation of the peripheral chemore-ceptors results in decreased reabsorption of renaltubular sodium (Honig 1989). Tissue hypoxia mayalso dilate renal vessels directly (Halliwill 2003),which would increase the glomerular filtration rate (Olsen et al. 1993) and thus sodium and waterexcretion. Several hormones may also play a keyrole in the fluid regulation at high altitude, since thenatriuretic and diuretic response is not abolished by renal nerve section (Schmidt et al. 1985; Karim et al. 1987). In fact, after adrenalectomy, the strikingincrease in urinary sodium excretion on exposure to hypobaric hypoxia is not observed (Lewis et al.1942).

Page 463: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 447

Metabolic

Upon acute exposure to altitude, carbohydratesmay be the preferred fuel since they have a highyield of adenosine triphosphate (ATP) per liter of O2consumed (Brooks et al. 1991; Roberts et al. 1996b). In support of this hypothesis, hypoxia inducible factor-1α (HIF-1α), which is known to increase thetranscription of several genes involved in glucoseuptake and glycolysis, is up-regulated upon acuteexposure to hypoxia (Semenza et al. 1994; Firth et al. 1995). In addition, lactate accumulation is increased during both rest and exercise upon acute exposureto altitude, which has been attributed to acceleratedglycolytic metabolism (Knuttgen & Saltin 1973).However, no differences in muscle glycogen utiliza-tion have been observed during exercise upon acuteexposure to exercise compared to sea level (Young,A.J. et al. 1982; Green et al. 1992). With chronic alti-tude exposure, both increased (Brooks et al. 1991;Roberts et al. 1996b) and decreased (Young, A.J. et al.1982, 1991; Beidleman et al. 1997; Braun et al. 2000)utilization of carbohydrates have been reported.However, lactate accumulation is consistently de-pressed despite a similar O2 delivery followingchronic altitude exposure (Young, A.J. et al. 1982;Brooks et al. 1991; Beidleman et al. 1997). This hasbeen termed the ‘lactate paradox’ and has beenattributed to decreased β-adrenergic stimulation ofmuscle glycogenolysis (Mazzeo & Reeves 2003).

Acute mountain sickness

Acute mountain sickness (AMS) is a syndromeinduced by hypoxia in unacclimatized individualswho ascend rapidly to altitudes exceeding 2500 mand remain there for more than several hours (Singhet al. 1969; Hackett & Rennie 1976; Honigman et al.1993). Characteristic symptoms include headache,nausea, vomiting, loss of appetite, fatigue, dizzinessand sleep disturbances (Hackett & Rennie 1976;Powles et al. 1978; Honigman et al. 1993; Sanchez del Rio & Moskowitz 1999; Bailey et al. 2000). Theonset of AMS usually occurs 4–12 h after ascent, andsymptoms become most prominent after the 1stnight spent at high altitude (Bärtsch et al. 1988). Theincidence of AMS increases with altitude; the

greater the hypoxemia, the more severe the illness(Maggiorini et al. 1990; Honigman et al. 1993).Individual AMS susceptibility can also be affectedby the rate of ascent (Hansen et al. 1967; Honigmanet al. 1993), prior acclimatization (Hansen et al. 1967;Lyons et al. 1995), obesity (Honigman et al. 1993) andlevel of exertion to reach a given altitude (Roach et al. 2000).

The pathophysiology of AMS is highly debatedbut in general is thought to be a vasogenic-inducedcerebral edema, triggered by the initial hypoxicstimulus (Hackett et al. 1998). Vasogenic edemacould be the result of increased hydrostatic pressureacross blood vessels due to an increase in cerebralblood flow or an altered permeability of the blood–brain barrier. Jensen et al. (1990) reported no increasein cerebral blood flow in subjects suffering fromAMS. However, increased expression of vascularendothelial growth factor has been reported inhumans (Walter et al. 2001) and animals (Xu &Severinghaus 1998) exposed to hypoxia or hypobarichypoxia, which would cause a generalized increasedcapillary leakage of large molecules due to endo-thelial activation in forming new blood vessels.

Resolution of AMS usually occurs following 1–2days of high altitude residence if no further ascent isattempted (Singh et al. 1969; Bärtsch et al. 1988).Physiological changes that may contribute to thisresolution of AMS include an increase in ventilationand diuresis, which can occur over the same fewdays of altitude residence (Singh et al. 1969; Hackettet al. 1982; Moore et al. 1986; Bärtsch et al. 1988). Theincrease in ventilation causes an increase in Pao2,which reduces the initial hypoxic stimulus. Anincreased diuresis also helps to reduce symptoms of AMS (Singh et al. 1969) via a reduction in extracel-lular fluid volume, which lessens the magnitude of cerebral edema, the causative factor for AMS(Hackett 1999).

Maximal exercise performance

According to the Fick equation, O2 uptake is theproduct of cardiac output (heart rate × stroke vol-ume) and the difference in Cao2 and venous O2 con-tent (Cvo2). Maximal Cao2 is decreased upon acuteexposure to altitude due to the reduction in ambient

Page 464: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

448 chapter 30

Po2 and corresponding reduction in Sao2 (Wolfel &Levine 2001). During maximal exercise at altitude,the body is unable to increase maximal ventilation,heart rate and tissue O2 extraction beyond maximalvalues obtained at sea level to compensate for thereduction in Sao2 (Stenberg et al. 1966; Grover et al.1986). Thus, maximal O2 uptake (Vo2max) is reducedin direct proportion to the decrease in maximal Cao2upon acute exposure to altitude (Stenberg et al. 1966;Vogel et al. 1967).

With chronic altitude exposure, maximal CaO2 isincreased compared to initial ascent due to increasesin both Sao2 and [Hb] (Wolfel & Levine 2001).Therefore, one would expect an increase in Vo2maxHowever, research has repeatedly shown thatVo2max does not change following chronic altitudeexposure (Saltin et al. 1968; Young, A.J. et al. 1982;Bender et al. 1988; Wolfel et al. 1991, 1998; Beidlemanet al. 1997). The reason for an unchanged Vo2max isthat maximal cardiac output decreases followingchronic altitude exposure due to a decrease in bothmaximal heart rate and stroke volume, which off-sets the increase in maximal Cao2 (Grover et al. 1986;Wolfel & Levine 2001). Thus, maximal O2 deliveryremains the same upon acute and chronic altitudeexposure (Grover et al. 1986).

Submaximal exercise performance

Oxygen uptake at rest or during submaximal fixedworkloads at altitude does not change from sealevel values (Reeves et al. 1967; Saltin et al. 1968;Raynaud et al. 1981). However, since Vo2max isdecreased at altitude, the relative exercise intensityduring a fixed submaximal workload can be greatlyincreased compared to that at sea level (Fig. 30.3). In this example, the same submaximal O2 uptake (2 L·min−1) represents 50% of a subject’s Vo2max atsea level and 70% of a subject’s Vo2max at altitude. Ifa fixed submaximal workload is maintained uponacute exposure to altitude, then ventilation, heartrate and tissue O2 extraction will be increased andsubmaximal exercise performance will be decreased(Ekblom et al. 1975; Fulco et al. 2003). If the submaxi-mal workload is adjusted such that an individual isworking at the same relative percentage of altitude-specific Vo2max, then ventilation, heart rate, tissue O2

extraction and submaximal exercise performancewill be similar at both sea level and during acuteexposure to altitude (Maher et al. 1974; Horstman et al. 1980).

With chronic altitude exposure, both small andlarge muscular endurance performance is improved,even when conducted at the same relative percent-age of altitude-specific Vo2max (Maher et al. 1974;Horstman et al. 1980; Fulco et al. 1994). Although notentirely understood, likely explanations for thisimprovement include a greater pulmonary diffu-sion time due to a decrease in blood flow (Boushel et al. 2001), a greater O2 diffusion gradient from themuscle capillary to the mitochondria (Fulco et al.1994; Beidleman et al. 2003) and sparing of muscleglycogen due to an increase in blood glucose util-ization (Brooks et al. 1991; Roberts et al. 1996a).Furthermore, AMS symptomatology is decreasedfollowing chronic altitude exposure which in and of itself may contribute to improvements in sub-maximal exercise performance.

Neurohumoral responses to altitude

Sympathoadrenal response

Plasma levels of norepinephrine (NE) are a functionof the rate of spillover into the circulation from sym-pathetic neuronal release (~ 70%), a small amount

Sea level

70%

4300 m

Steady-state O2 for cycle exerciseat power output of 100 W

5

4

3

2

1

0

Oxy

gen

up

take

(L·

min

–1)

50%

O2max

O2max O2max

Fig. 30.3 Effect of high altitude (4300 m) on the relationshipbetween absolute work load, oxygen uptake (Vo2), andrelative exercise intensity (% maximal oxygen uptake,Vo2max). (Modified from Young, A.J. & Young 1988.)

Page 465: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 449

secreted by the adrenal medulla (~ 30%), and its rate of removal from the plasma pool. Upon acuteexposure to normobaric hypoxia or hypobarichypoxia, numerous studies have reported an imme-diate increase in sympathetic nervous system (SNS)activity to skeletal muscle (Rowell et al. 1989;Leuenberger et al. 1991; Morgan et al. 1995) thatremains elevated following chronic altitude expos-ure (Hansen & Sander 2003). Although plasma NElevels do not always track SNS activity (Rowell et al.1989), summarized results from 30 studies also sug-gest that there is a 24% increase in resting NE levelsupon acute altitude exposure that increases a fur-ther 240% from acute to chronic altitude exposure(Fig. 30.4a). The mechanism for the initial and con-tinued elevation in SNS activity in hypoxia is notwell known, but is unlikely due to the direct effect ofhypoxia since SNS activity increases with time ataltitude, while hypoxemia decreases with time ataltitude (Mazzeo & Reeves 2003). It appears thathypoxia may cause sympathetic activation directlyvia stimulation of the peripheral chemoreceptors(Marshall 1994) and brainstem neuronal pools (Reiset al. 1994). In addition, unloading of cardiopul-monary baroreceptors could be involved due to adecrease in blood volume occurring over time at

altitude (Myhre et al. 1970; Jung et al. 1971; Jain et al.1980).

Similar to exercise at sea level, NE levels increaseone to threefold with exercise during acute andchronic altitude exposure (Mazzeo et al. 1991, 1995,2003; Young, A.J. et al. 1991), and the magnitude ofthe response is dependent on the intensity of exer-cise (Kjær et al. 1988; Braun et al. 2000; Mazzeo et al.2000; Lundby & Steensberg 2004). Summarizedresults suggest that the percentage of increase in NE during exercise from resting values is lower following chronic altitude exposure (~ 83%) com-pared to acute altitude exposure (~ 250%) and sealevel (~ 267%). However, NE levels reach higherabsolute levels during exercise with acute andchronic altitude exposure due to higher initial start-ing levels (Fig. 30.4a).

The immediate increase in SNS activity uponacute exposure to altitude likely plays a role incounterbalancing the effects of hypoxia-inducedvasodilation on most vascular beds so that bloodpressure can be maintained (Wolfel & Levine 2001).The rising NE levels during both rest and exercisefollowing chronic altitude exposure results inincreased arterial and venous tone, increased sys-temic vascular resistance, decreased plasma volume

SL(a) AA

AA

3.0

10.5

250

SL

2.4

8.8

267

CA

10.1

18.5

83

%SL-AA

24

20

%AA-CA

239

34

Rest

Exercise

% (R-Ex)

CA

RestExercise20

15

10

5

0

No

rep

inep

hri

ne

(nm

ol·L

–1)

SL(b)

AA

647

1840

184

SL

470

1040

121

CA

487

1226

152

%SL-AA

38

77

%AA-CA

–25

–33

Rest

Exercise

% (R-Ex)

CAAA

2000

1500

1000

500

0

Epin

eph

rin

e (p

mo

l·L–1

)

Fig. 30.4 Summarized results from 30 studies measuring (a) norepinephrine and (b) epinephrine levels during both restand exercise during sea level (SL), acute altitude (AA) and chronic altitude (CA) exposure (Kotchen et al. 1973b; Fulco et al. 1985; Bärtsch et al. 1988, 1991a; Bouissou et al. 1988, 1989; Férézou et al. 1988, 1993; Kjær et al. 1988; Richalet et al. 1988;Knudtzon et al. 1989b; Mazzeo et al. 1991, 1995, 2000, 2001, 2003; Young, A.J. et al. 1991, Olsen et al. 1992; Ramirez et al. 1992;Young, P.M. et al. 1992; Loeppky et al. 1993; Antezana et al. 1995; Larsen et al. 1997; Rostrup 1998; Sevre et al. 2001; Basu et al.2002; Beidleman et al. 2002; Bestle et al. 2002; Hasbak et al. 2002; Lundby & Steensberg 2004).

Page 466: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

450 chapter 30

and decreased cardiac output (Seals et al. 2001;Mazzeo & Reeves 2003). These physiologic adapta-tions serve to maintain a tight coupling betweenmetabolic supply and demand by decreasing bloodflow as arterial O2 delivery is improved followingchronic altitude exposure.

Sympathetic stimulation of the adrenal glandupon acute altitude exposure also causes an increasein plasma levels of epinephrine (EPI) that abates following chronic altitude exposure (Mazzeo &Reeves 2003). The summarized results from 30 stud-ies suggest that resting EPI levels increase 38% withacute altitude exposure and return to sea level val-ues following chronic altitude exposure (Fig. 30.4b).The primary mechanism for EPI release does appearto be hypoxia, since both the hypoxic stimulus and EPI levels are increased upon acute altitudeexposure and both lessen with chronic altitude resid-ence (Mazzeo & Reeves 2003). Wolfel et al. (1994)reported an inverse relationship between Sao2 andplasma EPI levels with time at altitude.

Similar to exercise at sea level, EPI levels increaseone to threefold with exercise from resting valuesduring both acute and chronic altitude exposure(Mazzeo et al. 1991, 1995, 2003; Young, A.J. et al.1991), and the magnitude of the response is depend-ent on the exercise intensity (Kjær et al. 1988; Braunet al. 2000; Mazzeo et al. 2000; Lundby & Steensberg2004). Summarized results suggest that the percent-age of increase in EPI during exercise from restingvalues is the same in all conditions (~ 120–180%),but reaches higher absolute levels with exercise during acute altitude exposure due to higher initialstarting levels (Fig. 30.4b). The increase and sub-sequent decrease in EPI levels during both rest andexercise likely contributes to the immediate increasein heart rate, vasodilation, muscle glycogenolysisand lactate accumulation upon acute altitude expos-ure that subsides following chronic exposure to altitude (Mazzeo & Reeves 2003). These physiologicadaptations serve to increase O2 delivery and optim-ize O2 utilization under limited O2 conditions andconserve cardiac energy as O2 supply improves.

Parasympathetic response

Several studies suggest a reduction in resting para-sympathetic nervous system (PNS) activity during

acute altitude exposure (Richardson et al. 1967; Kolleret al. 1988). In humans acutely exposed to hypoxia,pretreatment with propranolol (a β-adrenergicblocker) fails to prevent the acute increase in heartrate (Koller et al. 1988). However, when propranololand atropine (parasympathetic blocker) are com-bined, the increase in heart rate upon acute expos-ure to hypoxia is completely eliminated in bothhumans (Koller et al. 1988) and dogs (Hammill et al.1979). With chronic altitude exposure, most (Hartleyet al. 1974; Hughson et al. 1994; Boushel et al. 2001)but not all (Grover et al. 1986) studies suggest areversal of the initial decline in PNS activity suchthat cardiac vagal activity is increased and maximalheart rate decreased when compared to sea levelvalues. The mechanism for the initial decrease andsubsequent increase in PNS activity is not known,but may be induced via a baroreceptor-inducedexcitation of the vagal nerve in response to a slightdrop and subsequent elevation in mean arterialpressure with time at altitude (Hartley et al. 1974).The immediate decrease in PNS activity serves toincrease blood flow under limited O2 conditions byincreasing heart rate. The increase in PNS activityfollowing chronic altitude exposure may maintainmyocardial O2 demand, which is roughly estimatedfrom the rate-pressure product (heart rate × meanarterial pressure), by decreasing exercise heart rateas mean arterial pressure increases.

Glucocorticoid response

Cortisol (COR) is the major glucocorticoid pro-duced by the adrenal cortex and is secreted inresponse to increasing levels of adrenocorticotropichormone (ACTH) released from the anterior pitu-itary. Although individual study results may differ,the summarized results from 30 studies suggest that resting COR levels increase 33% with acute alti-tude exposure and return towards sea level valuesfollowing chronic altitude exposure (Fig. 30.5a). The trigger for the initial increase in ACTH andCOR with altitude exposure may be partly related to increased peripheral chemoreceptor input as aresult of the initial hypoxic stimulus, since deaf-ferentation of the arterial chemoreceptors results indiminished or abolished increases in ACTH andCOR (Raff et al. 1982; Honig et al. 1996).

Page 467: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 451

Similar to exercise at sea level, COR levelsincrease 30–100% with exercise during both acuteand chronic altitude exposure (Bouissou et al. 1988;Kjær et al. 1988; Bärtsch et al. 1991a; Braun et al.2000), and the magnitude of the response is depend-ent on the intensity of exercise (Kjær et al. 1988;Braun et al. 2000). Summarized results suggest thatthe percentage of increase in COR during exercisefrom resting values is the same in all conditions (~ 15–33%) but reaches higher absolute values with exercise during acute altitude exposure due to higher initial starting values (Fig. 30.5a). Theincrease in COR upon acute exposure to altitudeduring both rest and exercise most likely contributesto increased cardiac contractility, cardiac output,erythropoietin synthesis and energy mobilizationthrough gluconeogenesis (White et al. 1995).

Erythropoietin

Erythropoietin (EPO) is a glycoprotein hormonethat is released primarily from the kidneys and to a lesser degree by the liver. Summarized results

from 15 studies suggest that initial ascent to altitudeinduces a two to threefold increase in resting EPOlevels, with peak levels occurring within ~ 24–48 h,that declines toward but does not reach sea levelvalues following chronic altitude exposure (Fig.30.5b). The EPO response to exercise with acute and chronic altitude exposure has not been wellstudied. Thus, whether or not the EPO response ispotentiated during hypoxic exercise cannot bedetermined at this time. The initial stimulus for EPOproduction is related to the reduction in tissue O2tension in the renal cortex upon acute altitude expos-ure (Jelkmann 1992). The actual renal O2 sensor is probably a heme protein (Goldberg et al. 1998), or an enzyme termed hypoxia-inducible factor-αprolyl-hydroxylase (HIF-PH) (Ivan et al. 2001;Jaakkola et al. 2001). In the presence of decreased O2delivery, activation of this sensor appears to lead to the synthesis of HIF-1α, which then binds to theactive site on the enhancer region of the EPO geneand activates transcription (Semenza et al. 1991).This immediate increase in EPO stimulates thehematological cascade which eventually leads to

SL(a) AA

AA

470

541

15

SL

354

470

33

CA

388

484

25

%SL-AA

33

15

%AA-CA

–18

–10

Rest

Exercise

% (R-Ex)

CA

RestExercise

600

500

400

300

200

100

0

Co

rtis

ol (

nm

ol·L

–1)

AA

51

SL

13

CA

25

%SL-AA

292

%AA-CA

–51Rest

SL(b) CAAA

60

50

40

30

20

10

0

Eryt

hro

po

ieti

n (

IU·L

–1)

Fig. 30.5 Summarized results from 30 studies measuring (a) cortisol during both rest and exercise (Moncloa et al. 1968;Singh et al. 1974; Frayser et al. 1975; Sutton 1977; Sutton et al. 1977; Claybaugh et al. 1982; Heyes et al. 1982; Mordes et al.1983; Maresh et al. 1984; Bärtsch et al. 1988, 1991a; Bouissou et al. 1988; Friedl et al. 1988; Kjær et al. 1988; Richalet et al. 1989;Rock et al. 1989; Tunny et al. 1989; Kraemer et al. 1991; Sawhney et al. 1991; Ramirez et al. 1992; Vuolteenaho et al. 1992;Westendorp et al. 1993; Banfi et al. 1994, 1996; Beidleman et al. 1997, 2002; Larsen et al. 1997; Zaccaria et al. 1998; Braun et al.2000; Basu et al. 2002) and 15 studies measuring (b) erythropoietin (EPO) at rest during sea level (SL), acute altitude (AA)and chronic altitude (CA) exposure (Abbrecht & Littell 1972; Milledge & Cotes 1985; Eckardt et al. 1989; Mairbaurl et al.1990; Gunga et al. 1994; Klausen et al. 1996; Sawka et al. 1996; Gleiter et al. 1997; Chapman et al. 1998; Grover et al. 1998;Hudson et al. 1999; Bonfichi et al. 2000; Koistinen et al. 2000; Reeves et al. 2001; Ge et al. 2002).

Page 468: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

452 chapter 30

an increase in RBC production if the altitude sojournis prolonged.

The reason for the decrease in EPO to near sealevel values despite continuing hypoxemia is dif-ficult to explain. Just like any other hormone, thereis a dynamic equilibrium between production andconsumption. If increased erythroid activity follow-ing chronic altitude exposure leads to increased EPOconsumption, plasma levels of EPO may decline inthe face of elevated EPO turnover. Furthermore,there may be other stimuli that change follow-ing chronic altitude exposure such as increasing 2,3 diphosphoglycerate levels (Miller et al. 1973),decreasing sympathetic stimulation (Mazzeo &Reeves 2003) and down-regulation of HIF-1α(D’Angelo et al. 2003), which could explain declin-ing EPO levels despite persistent systemic hypoxia.

Fluid regulatory hormones

Renin–angiotensin–aldosterone system. Renin is a gly-coprotein released from the juxtaglomerular cells of

the kidney. Renin cleaves the circulating substrateangiotensinogen to angiotensin I, which is then further metabolized to its active form, angiotensinII, by angiotensin-converting enzyme in the lungvascular endothelium. Angiotensin II, a potentvasoconstrictor, stimulates secretion of aldosterone(ALDO) by the adrenal cortex. Renin is usually meas-ured by incubating the sample to be assayed andmeasuring the amount of angiotensin I generated.This measures the plasma renin activity (PRA) ofthe sample. Summarized results from 25 studiessuggest a 9% decrease in resting PRA levels uponacute altitude exposure that declines a further 10%from acute to chronic altitude exposure (Fig. 30.6a).The trigger for the slight and continued decrease inresting PRA and plasma renin with altitude expos-ure is also unknown, given that renin is typicallyreleased in response to β-adrenergic stimulationand decreasing blood pressure (Bouissou et al. 1989).Since β-adrenergic stimulation is increased withacute altitude exposure and decreased with chronicaltitude exposure, one would expect a parallel

SL(a) AA

AA

2.0

5.4

170

SL

2.2

5.5

150

CA

1.8

6.0

233

%SL-AA

–9

–2

%AA-CA

–10

11

Rest

Exercise

% (R-Ex)

CA

RestExercise7

6

5

4

3

2

1

0Plas

ma

ren

in a

ctiv

ity

(ng

·mL–1

h–1

)

AA

233

629

169

SL

326

897

175

CA

206

706

244

%SL-AA

–29

–30

%AA-CA

–10

12

Rest

Exercise

% (R-Ex)

SL(b) CAAA

1000

800

600

400

200

0

Ald

ost

ero

ne

(pm

ol·L

–1)

Fig. 30.6 Summarized results from 25 studies measuring (a) plasma renin activity during both rest and exercise (Kotchenet al. 1973a; Frayser et al. 1975; Maher et al. 1975; Sutton et al. 1977; Heyes et al. 1982; Milledge et al. 1983a; Bärtsch et al. 1988,1991a, 1991b; Bouissou et al. 1988, 1989; Knudtzon et al. 1989b; Tunny et al. 1989; De Angelis et al. 1992, 1996; Olsen et al.1992; Ramirez et al. 1992; Vuolteenaho et al. 1992; Rock et al. 1993; Westendorp et al. 1993; Antezana et al. 1995; Angelini et al. 1997; Zaccaria et al. 1998; Bestle et al. 2002; Robach et al. 2002) and 28 studies measuring (b) aldosterone during bothrest and exercise (Frayser et al. 1975; Maher et al. 1975; Sutton et al. 1977; Claybaugh et al. 1982; Heyes et al. 1982; Milledge et al. 1983a, 1989; Maresh et al. 1984; Bärtsch et al. 1988, 1991a, 1991b; Bouissou et al. 1988, 1989; Knudtzon et al. 1989b;Tunny et al. 1989; De Angelis et al. 1992, 1996; Olsen et al. 1992; Ramirez et al. 1992; Vuolteenaho et al. 1992; Loeppky et al.1993; Rock et al. 1993; Westendorp et al. 1993; Antezana et al. 1995; Angelini et al. 1997; Zaccaria et al. 1998; Bestle et al. 2002;Robach et al. 2002) during sea level (SL), acute altitude (AA) and chronic altitude (CA) exposure.

Page 469: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 453

increase and decrease in PRA and plasma renin. Thehypoxic suppression of PRA with acute and chronicaltitude exposure may be related to the increase in atrial natriuretic peptide with acute altitudeexposure (Johnston et al. 1989), or stimulation of anintrarenal baroreceptor due to increased renal per-fusion following chronic altitude exposure (Hoganet al. 1973; Olsen 1995).

Similar to exercise at sea level, PRA or plasmarenin levels increase one to threefold with exerciseduring both acute and chronic altitude exposure(Maher et al. 1975; Bouissou et al. 1988; Bärtsch et al.1991a; Rock et al. 1993). Summarized results suggestthat the percentage of increase in PRA or renin during exercise from resting values is the same in all conditions (~ 150–233%), and absolute levelsreached during exercise are also similar in all con-ditions (Fig. 30.6a).

Although suspected for some time, it is now generally agreed that the production of angiotensinII and activity of angiotensin-converting enzymeare not inhibited by hypoxia (Milledge & Catley1987; Raff et al. 1989). Thus, ALDO secretionremains under predominant control of the renin–angiotensin system even at altitude (Olsen 1995).Thus, it is not surprising that the summarizedresults of 28 studies suggest that resting levels ofALDO are also reduced 30–50% from sea level values upon acute and chronic altitude exposure,respectively (Fig. 30.6b). Although the most likelyreason for the decreased ALDO levels with acuteand chronic altitude exposure is the decreased reninvalues (Olsen 1995), a decrease in plasma K+ andACTH with altitude acclimatization may also con-tribute (Okazaki et al. 1984).

Similar to exercise at sea level, ALDO levelsincrease one to threefold with exercise during bothacute and chronic altitude exposure (Sutton 1977;Bouissou et al. 1988; Bärtsch et al. 1991a; Braun et al.2000; Beidleman et al. 2002). Summarized resultssuggest that the percentage of increase in ALDOduring exercise from resting values is the same in allconditions (~ 169–244%), but reaches lower abso-lute levels during acute and chronic altitude expos-ure due to lower initial starting levels (Fig. 30.6b).The decline in ALDO levels during both rest andexercise most likely contributes to the increased

natriuresis and diuresis upon acute and chronic altitude exposure. These physiological adaptationswould be beneficial for increasing [Hb] and O2transport as well as decreasing brain edema andthus AMS (Hackett 1999).

Atrial natriuretic peptide. Atrial natriuretic peptide(ANP) is a polypeptide released by the cardiac atrialmuscle fibers in response to overstretch of the atria.Although the literature is conflicting, summarizedresults of 21 studies suggest that plasma ANPincreases 26% upon acute altitude exposure andreturns to sea level values following chronic altitudeexposure (Fig. 30.7a). The initial stimulus for ANPrelease may be the hypoxic pulmonary vasocon-striction that occurs upon acute exposure to alti-tude, but does not explain the decrease in ANPgiven the sustained increase in pulmonary arter-ial pressure following chronic altitude exposure(Boussuges et al. 2000). Given the stimulatory effectsof HIF-1α on transactivation of ANP in hypoxicatrial myocytes (Chen, Y.F. et al. 1997), up- anddown-regulation of HIF-1α with acute and chronicaltitude exposure may explain increasing and de-creasing ANP levels (D’Angelo et al. 2003).

Similar to exercise at sea level, ANP levelsincrease 50–150% with exercise during both acuteand chronic altitude exposure (Bärtsch et al. 1991a;Olsen et al. 1992; Rock et al. 1993; Braun et al. 2000).Summarized results suggest that the percentage ofincrease in ANP during exercise is lower followingchronic altitude exposure (~ 3%) compared to sealevel (~ 88%) and acute altitude exposure (~ 72%)(Fig. 30.7a). The immediate increase in ANP likelycontributes to the increased diuresis and reductionin plasma volume observed upon acute altitudeexposure.

Antidiuretic hormone. Antidiuretic hormone (ADH)is a peptide hormone stored in the posterior pitu-itary and released in response to increased osmoticpressure. The summarized results of 16 studies suggest that ADH increases 10% with acute altitudeexposure and returns to sea level values follow-ing chronic altitude exposure (Fig. 30.7b). Due to the limited number of studies, the ADH response to exercise cannot be summarized. Bartsch and

Page 470: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

454 chapter 30

colleagues (Bärtsch et al. 1991a) reported the same percentage of increase in ADH during exerciseboth at sea level and upon acute altitude exposure,while Robach et al. (2002) found enhanced ADHrelease during exercise following chronic altitudeexposure. Clearly, further research needs to be con-ducted before definitive insight can be gained on the ADH response to exercise at altitude. The slightincrease in ADH upon acute altitude exposurecould be the result of an increase in peripheralchemoreceptor activity (Share & Levy 1966) orplasma osmolality (Bestle et al. 2002; Maresh et al.2004), but doesn’t explain the decrease in ADH aschemoreceptor activity (Bisgard & Forster 1996) andplasma osmolality (Blume et al. 1984; De Angelis et al. 1992; Bestle et al. 2002; Maresh et al. 2004)increase following chronic altitude exposure. Heyeset al. (1982) found that ADH levels were inverselycorrelated with blood pressure. Thus, the acute fallin blood pressure upon acute exposure to altitudemay stimulate an increase in ADH, while thechronic increase in blood pressure following alti-tude residence may cause ADH levels to return tonormal. Given that an increase in ADH would

oppose the beneficial effects of diuresis, the reasonfor the slight increase in ADH upon acute altitudeexposure is not clear.

Glucogregulatory hormones

Insulin. Insulin (INS) is a polypeptide hormonesecreted in response to high blood glucose levels bybeta cells in the islets of Langerhans. Summarizedresults from 13 studies suggest that plasma INSincreases 12% upon acute altitude exposure andreturns toward but does not reach sea level valuesfollowing chronic altitude exposure (Fig. 30.8a). Theinitial increase and subsequent decrease in INSsecretion with altitude exposure is not entirelyknown, since resting blood glucose levels are notaltered by acute altitude exposure (Young, P.M. et al. 1989b; Roberts et al. 1996b; Braun et al. 2001)and tend to decrease following chronic altitudeexposure (Stock et al. 1978; Young, P.M. et al. 1989b;Roberts et al. 1996b). The increase in INS with acutealtitude exposure may be due to β-adrenergic stimu-lation. The subsequent decrease in INS followingchronic altitude exposure may be due to rising NE

SL(a) AA

AA

14.7

25.3

72

SL

11.6

21.8

88

CA

11.6

12.0

3

%SL-AA

26

16

%AA-CA

–21

–53

Rest

Exercise

% (R-Ex)

CA

RestExercise

25

20

15

10

5

0Atr

ial n

atri

ure

tic

pep

tid

e (p

mo

l·L–1

)

AA

2.6

SL

2.3

CA

2.3

%SL-AA

10

%AA-CA

–10Rest

SL(b) CAAA

3.0

2.5

2.0

1.5

1.0

0.5

0.0

An

tid

iure

tic

ho

rmo

ne

(pm

ol·L

–1)

Fig. 30.7 Summarized results from 21 studies measuring (a) atrial natriuretic peptide during both rest and exercise(Bärtsch et al. 1988, 1991a, 1991b; Bouissou et al. 1989; Kawashima et al. 1989; Milledge et al. 1989; Tunny et al. 1989; Koller et al. 1991; De Angelis et al. 1992, 1996; Olsen et al. 1992; Ramirez et al. 1992; Vuolteenaho et al. 1992; Loeppky et al. 1993;Rock et al. 1993; Westendorp et al. 1993; Antezana et al. 1995; Rostrup 1998; Zaccaria et al. 1998; Bestle et al. 2002; Robach et al. 2002) and 16 studies measuring (b) antidiuretic hormone at rest (Singh et al. 1974; Claybaugh et al. 1982; Heyes et al.1982; Blume et al. 1984; Porchet et al. 1984; Bärtsch et al. 1988, 1991a, 1991b; Koller et al. 1991; De Angelis et al. 1992, 1996;Loeppky et al. 1993; Rostrup 1998; Sevre et al. 2001; Robach et al. 2002; Maresh et al. 2004) during sea level (SL), acutealtitude (AA) and chronic altitude (CA) exposure.

Page 471: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine

hormones and altitude 455

levels, which would suppress INS release via en-hanced α2-adrenergic stimulation (Schmidt et al.1997; Schäfers et al. 1999).

Similar to exercise at sea level, INS levels decreasefrom 10% to 50% with exercise during both acuteand chronic altitude exposure (Sutton 1977; Brookset al. 1991; Young, P.M. et al. 1992; Braun et al. 2000;Beidleman et al. 2002). Summarized results sug-gest that the percentage of decrease in INS duringexercise from resting values is slightly less follow-ing chronic altitude exposure (~ 19%) compared to sea level (–39%) or acute altitude exposure (–49%) (Fig. 30.8a). However, if the studies wereconducted at the same relative intensity of exerciseat both sea level and altitude, the percentage ofdecline in INS during exercise is similar (Braun et al. 2000; Beidleman et al. 2002). The increased INSlevels upon acute altitude exposure may contributeto the preferential use of carbohydrates in order to maximize O2 fuel efficiency under limited O2conditions.

Glucagon. Glucagon (GLG) is also a polypeptidehormone secreted in response to low blood glucoselevels by α cells in the Islets of Langerhans. Since

summarized results would be skewed by the lownumber of GLG measurements at altitude, individ-ual study results will be examined. Given the anti-insulin properties of GLG, one would expect a slightdecrease upon acute altitude exposure that returnstoward normal sea level values following chronicaltitude exposure. Most (Kjær et al. 1988; Roberts et al. 1996b; Larsen et al. 1997) but not all (Beidlemanet al. 2002) studies have reported a slight 5%decrease in GLG upon acute exposure to altitude.This slight decrease in GLG may facilitate enhancedglycolysis. However, only two studies have ex-amined the resting GLG response following chronicaltitude exposure, and one reported a 55% increase(Roberts et al. 1996b) while the other reported nochange (Larsen et al. 1997). Thus, whether or notGLG changes following chronic altitude exposureremains unknown.

Similar to exercise at sea level, GLG increases10–30% during exercise at altitude (Roberts et al.1996b; Beidleman et al. 2002), which is most likelyrelated to the exercise-induced depletion of glucose.It does not appear that the GLG response to exerciseis potentiated by altitude exposure, but furtherresearch is needed in this area.

SL(a) AA

AA

89

45

49

SL

79

48

39

CA

83

67

19

%SL-AA

12

–6

%AA-CA

–7

48

Rest

Exercise

% (R-Ex)

CA

RestExercise

100

80

60

40

20

0

14

12

10

8

6

4

2

0

Insu

lin (

pm

ol·L

–1)

AA

2.7

11.7

333

SL

3.0

8.8

193

CA

2.1

7.8

271

%SL-AA

–8

10

%AA-CA

–22

–33

Rest

Exercise

% (R-Ex)

SL(b) CAAA

Gro

wth

ho

rmo

ne

(ng

·mL–1

)

Fig. 30.8 Summarized results from 13 studies measuring (a) insulin during both rest and exercise (Sutton 1977; Stock et al.1978; Sawhney et al. 1986; Young, P.M. et al. 1989a, 1992; Brooks et al. 1991; Sawhney et al. 1991; Young, A.J. et al. 1991;Roberts et al. 1996b; Larsen et al. 1997; Braun et al. 2000, 2001; Beidleman et al. 2002) and nine studies measuring (b) growthhormone during rest (Sutton et al. 1970; Sutton 1977; Raynaud et al. 1981; Kjær et al. 1988; Knudtzon et al. 1989a; Sawhney et al. 1991; Banfi et al. 1994; Larsen et al. 1997; Beidleman et al. 2002) during sea level (SL), acute altitude (AA) and chronicaltitude (CA) exposure.

Page 472: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 473: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 474: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 475: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 476: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 477: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 478: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 479: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 480: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 481: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 482: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 483: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 484: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 485: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 486: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 487: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 488: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 489: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 490: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 491: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 492: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 493: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 494: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 495: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 496: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 497: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 498: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 499: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 500: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 501: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 502: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 503: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 504: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 505: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 506: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 507: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 508: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 509: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 510: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 511: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 512: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 513: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 514: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 515: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 516: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 517: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 518: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 519: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 520: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 521: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 522: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 523: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 524: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 525: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 526: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 527: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 528: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 529: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 530: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 531: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 532: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 533: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 534: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 535: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 536: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 537: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 538: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 539: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 540: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 541: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 542: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 543: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 544: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 545: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 546: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 547: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 548: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 549: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 550: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 551: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 552: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 553: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 554: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 555: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 556: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 557: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 558: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 559: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 560: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 561: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 562: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 563: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 564: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 565: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 566: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 567: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 568: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 569: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 570: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 571: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 572: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 573: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 574: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 575: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 576: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 577: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 578: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 579: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 580: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 581: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 582: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 583: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 584: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 585: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 586: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 587: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 588: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 589: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 590: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 591: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 592: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 593: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 594: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 595: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 596: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 597: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 598: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 599: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 600: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 601: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 602: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 603: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 604: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 605: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 606: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 607: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 608: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 609: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 610: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 611: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 612: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 613: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 614: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 615: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 616: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 617: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 618: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 619: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 620: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 621: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 622: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 623: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 624: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 625: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 626: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 627: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 628: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 629: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 630: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 631: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 632: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 633: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 634: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 635: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 636: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 637: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 638: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 639: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 640: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 641: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 642: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 643: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 644: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 645: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine
Page 646: THE ENDOCRINE SYSTEM IN SPORTS AND EXERCISE Library...The endocrine system in sports and exercise / edited by W.J. Kraemer and A.D. Rogol. p. ; cm. a (The encyclopaedia of sports medicine