myocyte androgen receptor modulates body …...myocyte androgen receptor modulates body composition...

195
MYOCYTE ANDROGEN RECEPTOR MODULATES BODY COMPOSITION AND METABOLIC PARAMETERS by Shannon M. Fernando Hon. B.Sc. University of Toronto, 2008 A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Shannon M. Fernando, 2010 UNIVERSITY OF TORONTO July, 2010 All rights reserved. This work may not be reproduced in whole or in part, by photocopy or other means, without the permission of the author

Upload: others

Post on 26-Mar-2020

11 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

MYOCYTE ANDROGEN RECEPTOR MODULATES BODY COMPOSITION AND

METABOLIC PARAMETERS

by

Shannon M. Fernando

Hon. B.Sc. University of Toronto, 2008

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Institute of Medical Science

University of Toronto

© Shannon M. Fernando, 2010

UNIVERSITY OF TORONTO

July, 2010

All rights reserved. This work may not be

reproduced in whole or in part, by photocopy or other means,

without the permission of the author

Page 2: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

ii

Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute of Medical Science, University of Toronto

ABSTRACT

Androgens (such as testosterone) have been shown to increase lean body mass and

reduce fat body mass in men through activation of androgen receptors (AR). While this

suggests a potential clinical use for androgens, attempts at utilization of this class of

hormones as a therapeutic are limited by side effects due to indiscriminate AR activation

in various tissues. Thus, a greater understanding of the tissues and cells involved in

promoting these changes would be beneficial. Here we show that selective overexpression

of AR in muscle cells of transgenic (HSA-AR) rodents both increases lean muscle mass

and significantly reduces fat mass in males. Similar effects can be induced in HSA-AR

females treated with testosterone. Metabolic analyses of HSA-AR males show that these

animals demonstrate increased O2 consumption and hypermetabolism. Thus, targeted

activation of AR in muscle regulates body composition and metabolism, suggesting a

novel target for drug development.

Page 3: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

iii

ACKNOWLEDGEMENTS

None of the work contained within these pages could have been accomplished

without the help of many wonderful people. I must begin by thanking my supervisor, Dr.

Ashley Monks, for his incredible guidance and support over the course of my time in his

laboratory. I entered his lab as a very “green” undergraduate, but his knowledge and trust

have allowed me the opportunity to learn the pitfalls and plateaus of science. He took a

chance on me, and for that I am forever grateful. I have thoroughly enjoyed my time

under his tutelage, as well as our many collaborative efforts, and I can only hope that I

have given back as much as he has given to me. I also thank the members of my Program

Advisory Committee, Drs. Angela Lange and Tim Westwood, for their input and

encouragement. I thank the members of the Monks Lab for their consistent help and

support. Most notably, I must acknowledge the efforts of Dr. Lee Niel, who seemingly

helped me in everything I did from the first moment I walked into the lab. I also thank Dr.

Pengcheng Rao, Dr. Melissa Holmes, Dr. Kaiguo Mo, Dr. Diptendu Chatterjee, Marijana

Stagljar and Mutaz Musa for technical support. The learning curve would have been

much steeper if not for you all.

I am very grateful to have the unconditional love and support of Arthur, Jini and

Krystyna Fernando. Many thanks for putting up with my complaints, and for

understanding the importance of the words: “I have work to do.”

Lastly, I must acknowledge the Natural Sciences and Engineering Research

Council of Canada, the Ontario Graduate Scholarship Program, and the University of

Toronto Open Fellowships for the funding of my research.

Page 4: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

iv

TABLE OF CONTENTS

TITLE PAGE..................................................................................................................... I ABSTRACT ...................................................................................................................... IIACKNOWLEDGEMENTS ........................................................................................... IIILIST OF TABLES......................................................................................................... VIILIST OF FIGURES..................................................................................................... VIIICHAPTER 1: INTRODUCTION .................................................................................... 11.1.OVERVIEW..........................................................................................................................11.2.ANINTRODUCTIONTOBODYCOMPOSITION ....................................................................21.2.1.Lean(Muscle)Mass ..................................................................................................................21.2.2.FatMass.........................................................................................................................................61.2.3.Bone.................................................................................................................................................8

1.3.ENERGYBALANCEANDMETABOLISM............................................................................. 101.3.1.WhatisEnergyBalance?..................................................................................................... 101.3.2.EnergyIntake ........................................................................................................................... 121.3.3.EnergyExpenditure ............................................................................................................... 151.3.4.EnergyBalance,BodyCompositionandObesity....................................................... 18

1.4.THEANDROGENSYSTEM ................................................................................................ 211.4.1.AndrogenPharmacology–StructureandProduction........................................... 221.4.2.AndrogenReceptor ................................................................................................................ 25

1.5.ANDROGENS,ARANDMUSCLE....................................................................................... 271.5.1.AnabolicFunctionsofARinMuscle................................................................................ 281.5.2.MuscleARandNeuromuscularDevelopment ............................................................ 311.5.3.SpinalandBulbarMuscularAtrophy ............................................................................ 34

1.6.ANDROGENS,ARANDFAT ............................................................................................. 361.6.1.SexDifferencesinAdiposity ............................................................................................... 361.6.2.AndrogensandAdipocyteDevelopment....................................................................... 40

1.7.WHERETHEACTIONIS:LESSONSLEARNEDFROMARKO ........................................... 421.7.1.Whole­BodyAndrogenReceptorKnockout................................................................. 441.7.2.Tissue­SpecificAndrogenReceptorKnockout............................................................ 46

1.8.THEMETABOLICPOTENTIALOFMUSCLE ...................................................................... 491.8.1.SkeletalMuscleMetabolism............................................................................................... 491.8.2.MuscleMitochondria............................................................................................................. 53

1.9.OBJECTIVESANDHYPOTHESES ....................................................................................... 58CHAPTER 2: MATERIALS AND METHODS ........................................................... 602.1.OVERVIEW....................................................................................................................... 602.2.ANIMALSTRAINS............................................................................................................. 602.2.1.HSA­ARRats.............................................................................................................................. 602.2.2.TfmRats...................................................................................................................................... 622.2.3.HSA­AR/TfmRats ................................................................................................................... 642.2.4.HSA­ARMice ............................................................................................................................. 65

Page 5: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

v

2.3.SUBJECTS ......................................................................................................................... 672.3.1.Animals........................................................................................................................................ 672.3.2.Genotyping................................................................................................................................. 67

2.4.EXPERIMENTI:TISSUESPECIFICITYOFTRANSGENEEXPRESSION ................................ 692.4.1.Reverse­Transcription(End­Point)PCR....................................................................... 70

2.5.EXPERIMENTII:BODYCOMPOSITIONANALYSIS ........................................................... 712.5.1.Dual­EnergyX­RayAbsorptiometry............................................................................... 712.5.2.ParametersExaminedandDerived................................................................................ 722.5.3.DissectionsandTissueWeights........................................................................................ 73

2.6.EXPERIMENTIII:TESTOSTERONETREATMENTOFADULTFEMALES ........................... 732.6.1.T­CapsuleSurgeries ............................................................................................................... 742.6.2.BodyCompositionAnalysis................................................................................................. 75

2.7.EXPERIMENTIV:ADIPOSEHISTOLOGY.......................................................................... 752.7.1.SamplePreparationandSamplingStrategy.............................................................. 752.7.2.Measurements .......................................................................................................................... 77

2.8.EXPERIMENTV:ENERGYBALANCEANDMETABOLICANALYSES................................... 772.8.1.RestingMetabolismbyIndirectCalorimetry.............................................................. 772.8.2.SpontaneousActivityMeasures........................................................................................ 79

2.9.STATISTICALANALYSES .................................................................................................. 79CHAPTER 3: RESULTS ................................................................................................ 813.1.OVERVIEW....................................................................................................................... 813.2.EXPERIMENTI:TISSUESPECIFICITYOFTRANSGENEEXPRESSION ................................ 813.2.1.TransgenemRNAisExpressedinMuscleTissueofHSA­ARAnimals .............. 82

3.3.EXPERIMENTII:BODYCOMPOSITIONANALYSIS ........................................................... 823.3.1.HSA­ARExpressionDoesNotRegulateBodyMassinRats .................................. 833.3.2.IncreasedLeanMuscleMassPercentinHSA­ARMaleRats ................................ 833.3.3.ReducedFatBodyMassinHSA­ARMaleRats ........................................................... 853.3.4.NoEffectsonBodyCompositionofHSA­ARFemaleRats ..................................... 863.3.5.IndividualMuscleandFatPadWeights ....................................................................... 873.3.6.HSA­ARSimilarlyAffectsBodyCompositioninL78Mice ..................................... 883.3.7.EffectsofHSA­ARandTfmonBoneContentandDensity .................................... 89

3.4.EXPERIMENTIII:TESTOSTERONETREATMENTOFADULTFEMALERATS ................... 913.4.1.T­TreatmentRegulatesBodyCompositionofHSA­ARFemales......................... 923.4.2.T­TreatmentDoesNotAffectBodyCompositionofWild­typeFemales ......... 94

3.5.EXPERIMENTIV:ADIPOSEHISTOLOGY.......................................................................... 943.5.1.HSA­ARExpressionReducesAdipocyteArea.............................................................. 94

3.6.EXPERIMENTV:ENERGYBALANCEANDMETABOLICANALYSES................................... 953.6.1.HSA­ARExpressionIncreasesRestingMetabolisminRatsandMice .............. 963.6.2.SpontaneousActivityisNotAffectedbytheTransgene ........................................ 97

3.7.SUMMARY ........................................................................................................................ 97CHAPTER 4: DISCUSSION.......................................................................................... 994.1.OVERVIEW....................................................................................................................... 994.2.MYOCYTEARANDBODYCOMPOSITION......................................................................1004.2.1.LeanMuscleMass .................................................................................................................1014.2.2.AdiposeTissue........................................................................................................................104

Page 6: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

vi

4.2.3.Bone............................................................................................................................................1084.3.THEEFFECTOFTFMONBODYCOMPOSITION..............................................................1114.3.1.ComparisonwithARKOMice...........................................................................................1114.3.2.WhytheDiscrepancy?.........................................................................................................112

4.4.INTERACTIONSWITHESTROGENS.................................................................................1154.4.1.EstrogensandAdiposeTissue .........................................................................................1164.4.2.Estrogens,HSA­ARandTfm.............................................................................................117

4.5.MYOCYTEARANDOXIDATIVEMETABOLISM ..............................................................1204.5.1.LocalEffectsonSkeletalMuscleMetabolism ...........................................................1204.5.2.MitochondrialBiogenesisandEnzymeActivity ......................................................1214.5.3.EffectsonOtherMetabolicOrgans ...............................................................................124

4.6.COMPARINGHSA­ARRATSANDMICE........................................................................1264.6.1.PhenotypeComparisons ....................................................................................................1274.6.2.DoesExpressionLevelExaggeratePhenotype?.......................................................128

4.7.ANEWHOPE:SELECTIVEANDROGENRECEPTORMODULATORS ...............................1304.7.1.SelectiveAndrogenReceptorModulators..................................................................1314.7.2.TargetingMuscle ..................................................................................................................132

4.8.FUTUREDIRECTIONS ....................................................................................................1334.8.1.OtherMetabolicParametersinHSA­ARAnimals...................................................1334.8.2.IdentifyingARandMitochondrialInteractionsinSkeletalMuscle ................1354.8.3.MolecularandBiochemicalAssaysofKeyMetabolicPlayers...........................137

4.9.CONCLUSIONS ................................................................................................................139REFERENCES .............................................................................................................. 141TABLES AND FIGURES ............................................................................................. 161APPENDIX A: STASTICAL VALUES ...................................................................... 174

Page 7: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

vii

List of Tables

Table 1: Summary of animals used in each experiment

Table 2: List of primers used

Page 8: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

viii

List of Figures

Figure 1: Characterization of Transgene Expression

Figure 2: Transgene Expression Regulates Body Composition in Males

Figure 3: No Effect of the Transgene on Body Composition in Female Rats

Figure 4: Excised Fat Pads and Muscles Confirm DXA Findings

Figure 5: Body Composition of HSA-AR Mice

Figure 6: Effects of HSA-AR on Bone Parameters

Figure 7: Body Composition is Altered by T-treatment of HSA-AR Females

Figure 8: Smaller Adipocytes are Found in HSA-AR Males

Figure 9: Increased Oxygen Consumption in HSA-AR Male Rats

Figure 10: Differences in Energy Expenditure in HSA-AR L78 Mice

Figure 11: HSA-AR Expression Does Not Affect Spontaneous Activity Level

Page 9: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

ix

List of Abbreviations

aARKO = adipocyte-specific androgen receptor knockout AICAR = aminoimidazole carboxamide ribonucleotide ALS = amyotrophic lateral sclerosis AMPK = 5’ adenosine monophosphate-activated protein kinase αMSH = α-melanocyte stimulating hormone ANOVA = analysis of variance AR = androgen receptor ARKO = androgen receptor knockout ArKO = aromatase knockout AT = anterior tibialis ATP = adenosine triphosphate AVPV = anteroventral periventricular nucleus BAT = brown adipose tissue BC = bulbocavernosus BMC = bone mineral content BMD = bone mineral density BMR = basal metabolic rate CART = cocaine and amphetamine related transcript CP = creatine phosphate cDNA = complementary DNA C/EBP = CCAAT/enhancer binding protein α CNTF = ciliary neurotrophic factor DBD = DNA-binding domain DEPC = diethyl-pyrocarbonate DHT = dihydrotestosterone DNA = deoxyribonucleic acid DXA = dual-energy X-ray absorptiometry EDL = extensor digitorum longus ETC = electron transport chain FATP = fatty-acid transport protein FBM = fat body mass FSH = follicle-stimulating hormone GAPDH = glyceraldehyde-3-phosphate dehydrogenase GLUT4 = glucose transporter 4 GnRH = gonadotropin-releasing hormone GS = glycogen synthase HD = Huntington’s Disease HKII = hexokinase II HPG = hypothalamic-pituitary-gonadal HSA = human skeletal alpha-actin HSA-AR = human skeletal alpha-actin promoter, androgen receptor transgene IGF-1 = insulin-like growth factor-1 IHC = immunohistochemistry IR = insulin receptor

Page 10: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

x

kb = kilobase LA = levator ani LBM = lean body mass LH = luteinizing hormone LPL = lipoprotein lipase mARKO = myocyte-specific androgen receptor knockout MC4R = melanocortin 4 receptor MePD = posterodorsal medial amygdala mtDNA = mitochondrial DNA mRNA = messenger ribonucleic acid NPY = neuropeptide Y NTD = amino-terminal transactivation domain O2 = oxygen OCT = optimal cutting temperature embedding medium OD = optical density PAS = periodic acid-schiff PASD = diastase-digested periodic acid-schiff PBF = phosphate buffered formalin PBS = phosphate buffered saline PCOS = polycystic ovarian syndrome PCR = polymerase chain reaction PGC-1α = peroxisome proliferator-activated receptor gamma, coactivator 1 alpha PM = peritubular myoid PND = post-natal day POMC = proopiomelanocortin PPAR = peroxisome proliferator-activated receptor RMR = resting metabolic rate RNA = ribonucleic acid RT-PCR = reverse-transcription polymerase chain reaction SARM = selective androgen receptor modulator SBMA = spinal and bulbar muscular atrophy SCARKO = sertoli cell-specific androgen receptor knockout SD = Sprague-Dawley SDN-POA = sexually dimorphic nucleus of the preoptic area SHBG = sex hormone-binding globulin T = testosterone T2DM = type 2 diabetes mellitus Tfm = Testicular feminization mutation Tg = transgenic Type I = slow-twitch muscle fibers Type IIa = fast-twitch, oxidative muscle fibers Type IIb = fast-twitch, glycolytic muscle fibers UCP = uncoupling protein VEGF = vascular endothelial growth factor WAT = white adipose tissue WT = wild-type

Page 11: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

1

Chapter 1: Introduction 1.1. Overview

Perhaps the most well known function of androgens is its anabolic role in

regulating skeletal muscle mass. In men, this increase in skeletal muscle mass is largely

believed to be associated with activation of the androgen receptor (AR). However, men

who take exogenous amounts of androgens also experience a decrease in adipose tissue.

Significantly less is known about how androgens are capable of regulating body fat, and

improving body composition. Furthermore, significant controversy exists as to which

tissues possess the AR necessary for achieving these profound effects on body

composition. The principle purpose of this study was to investigate transgenic

(Tg) rats that overexpress AR in muscle fibers, and how this increased expression

contributes to changes in overall body composition and energy balance.

This introductory section provides the reader first with a general review of body

composition and the complex nature of energy homeostasis. This is followed by an

overview of the androgen system. A review of androgen function will be provided,

primarily focused upon how androgens are currently known to regulate muscle mass and

adipose tissue. Finally, discussion will turn toward the current problems in defining

androgen site-of-action in mediating body composition, as well as the metabolic potential

of skeletal muscle and the role it may play in regulating these effects. Finally, objectives

and hypotheses of this study will be described.

Page 12: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

2

1.2. An Introduction To Body Composition

Most (if not all) mammalian species on the planet share a common set of

components that constitute their collective mass (Corva & Medrano, 2000). These

common components provide a basis for homeostatic maintenance, diverse behaviours,

and development across the lifespan. Scientific study has largely separated these

components into three major types: lean (muscle) mass, fat mass, and bone. All three

serve important roles in the maintenance of mammalian life, and modulation of overall

body composition can have important implications for health and disease. Here we review

these major elements, understanding not only that each component of body composition

differs from others, but also the complexity contained within each.

1.2.1. Lean (Muscle) Mass

While the term “lean mass” is most accurately defined as the collection of non-fat,

non-bone tissue in the mammalian organism, many instead simply define it as overall

muscle mass. Muscle is the major type of contractile tissue contained within animals, and

is derived from the mesoderm layer during development (Orallo, 1996). It represents a

major site of nutrient metabolism, and functions through the movements of individual

contractile filaments that move past each other, subsequently changing the size of the

overall cell. There are three major types of muscle: smooth, cardiac, and skeletal.

The three types of muscle differ in various properties, including structure,

contractile properties, and control mechanisms (Orallo, 1996; Caplice & Deb, 2004;

Allen, Lamb & Westerblad, 2008). Smooth muscle is largely organized into sheet-like

structures and surrounds various hollow organs and tubes, including the stomach,

Page 13: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

3

intestines, urinary bladder, uterus, blood vessels, and airways in the lungs. It is non-

striated, and contraction of smooth muscle surrounding hollow organs may allow for the

propelling of luminal contents through the organ, or changes in the diameter of the tube.

Smooth muscle myocytes are attached to the hairs of the skin, as well as the iris of the

eye. Control of smooth muscle contraction is involuntary, and dictated by the autonomic

nervous system, endocrine/autocrine/paracrine agents, and other local chemical signals. In

contrast, cardiac muscle is a striated type of muscle that makes up the muscle of the heart.

Contraction of cardiomyocytes propels blood through the circulatory system. Similar to

smooth muscle, its contraction is involuntary, and under the control of similar neuronal

and endocrine signals. Finally, as its name suggests, skeletal muscle is attached to bone,

and is found throughout the body, from the rectus femoris of the leg to the trapezius of the

neck. Contraction of skeletal muscle is responsible for support and movement of the

skeleton. Similar to cardiac muscle, skeletal muscle is striated. However, unlike the other

types of muscle, skeletal muscle contraction occurs under voluntary control, and is

initiated by impulses in the neurons that innervate these muscles.

When studying body composition, skeletal muscle is often the most discussed

muscle-type, due to the fact that it is the most prone to change over the course of the

lifespan. Growth of skeletal muscle is typically associated with resistance exercise, a

prominent method used for achieving change in body composition. Similarly, skeletal

muscle is one of the most metabolically demanding tissues in mammalian bodies, and

thus changes in overall skeletal muscle weight can result in modulation of systemic

metabolism.

Page 14: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

4

Differences in skeletal muscle fiber types can be elucidated based upon

histochemical staining properties of the myosin adenosine triphosphatase (ATPase)

enzyme. Functional characteristics of each muscle fiber type are based upon its ATPase

enzyme activity. In this way, three major skeletal muscle fiber types have been found:

type I (slow-twitch), type IIa (fast-twitch, oxidative), and type IIb (fast-twitch, glycolytic)

(Staron, 1997). These fiber types can be distinguished on the basis of contraction time,

activity/duration of use, endurance, mitochondrial density, power, and

oxidative/glycolytic capacity. Type I (slow-twitch) fibers are predominantly used for

aerobic activity (Coen et al., 2010). As the name indicates, they have a slow contraction

time, and are characterized by a low activity level of ATPase. They contain large and

numerous mitochondria, and thus, are relatively resistant to fatigue. In contrast, type II

(fast-twitch) fibers demonstrate a significantly higher level of ATPase activity (Klover,

Chen, Zhu & Hennighausen, 2009). They have a much faster contraction time, and are

used during short-term, anaerobic activity. Type II fibers contain much less mitochondria

than type I fibers, and as a result, are far less resistant to fatigue. Type II fibers can also

be distinguished based upon their methods of energy utilization and metabolism. Certain

fast-twitch fibers are similar to slow-twitch fibers in that they strongly rely upon

oxidative metabolism for generation of ATP. These fibers are termed type IIa (fast-twitch,

oxidative). Conversely, type IIb fibers rely primarily upon anaerobic glycolysis for

generation of ATP. This method of energy generation is less efficient than oxidative

metabolism (less ATP per molecule of glucose), but is also faster. These fibers are thus

termed as fast-twitch, glycolytic fibers. Due to their reliance upon oxidative metabolism,

type IIa fibers tend to have relatively slower contraction time than type IIb fibers, but are

Page 15: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

5

also more resistance to fatigue. Individual muscles typically contain all three fiber types,

although the proportions of certain types are greater in specific muscles. For example, the

gastrocnemius muscle is largely composed of type II fibers, while the soleus muscle

(which together with the gastrocnemius constitutes the calf muscle) is largely composed

of type I fibers (Wang et al., 2004). Thus, even muscle groups in close proximity can be

composed of very different fiber type proportions.

Muscle fiber types can adapt to changing demands by altering size or fiber type

composition (Scott, Stevens & Binder-MacLeod, 2001). This is typically what occurs

when individuals take part in exercise training, whether resistance- or endurance-based.

Both types of training tend to result in muscle fiber hypertrophy, although there is some

disagreement as to which fiber types are preferentially altered by exercise (Adams,

Hather, Baldwin & Dudley, 1993). Both endurance and resistance training result in

similar reductions of MHC coexpression, resulting in a greater number of “pure” fibers.

However, while both types of exercise result in similar trends in fiber type conversion, the

physiological changes induced by each type of exercise are rather different. Endurance

training results in an increase of muscle oxidative capacity (hypertrophy of type I fibers),

while resistance training increases overall muscle fiber strength and size (hypertrophy of

type II fibers).

Muscular contraction is a major energetically demanding process. Contraction of

skeletal muscle mediates all voluntary movement and physical action in mammalian

organisms. The ability of a muscle fiber to generate force and movement depends on the

interaction of the two major contractile proteins: actin and myosin (Kee, Gunning &

Hardeman, 2009). Actin molecules are globular proteins composed of a single

Page 16: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

6

polypeptide, and polymerize with other actins in order to form intertwined helical chains.

These chains make up the core of a thin filament in the sarcomere. Myosin, conversely, is

composed of two large polypeptide heavy chains, and four smaller light chains. These

polypeptides combine to form a molecule that consists of two globular heads, and a long

tail. The tail of each myosin molecule lies along the axis of the thick filament, and the

two globular heads extend out to the sides, forming the cross-bridges. Each globular head

contains two binding sites, one for actin, and one for ATP. During contraction, ATP is

hydrolyzed (to form ADP and a phosphate group). Using the energy from ATP

hydrolysis, myosin is able to bind to actin, and pull adjacent actin molecules together.

After the end of a contraction sequence, the binding of a new ATP molecule is required in

order for the link between actin and myosin to be broken. Thus, one can see that muscle

contraction requires high amounts of energy in the form of ATP.

1.2.2. Fat Mass

Triglyceride (fat) consists of three fatty acids, which are linked to a glycerol

group. Fat accounts for approximately 80 percent of the energy that is stored in the body

(Friedman, 2009). Under typical resting conditions, approximately half of the energy

utilized by muscle, liver and kidney is derived from catabolism of fatty acids. In

mammalian systems, although most cells do store some small amounts of fat, the

predominant localization of these molecules is in adipose tissue (Rosen & Spiegelman,

2000). This is a collection of loose connective tissue composed of individual fat cells

called adipocytes. Adipose tissue is largely localized to specific depots within the body. It

either exists in various deposits underlying the skin (termed ‘subcutaneous fat’) or in

Page 17: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

7

larger aggregates in body cavities such as the abdomen (termed ‘visceral fat’). The

function of individual adipocytes is to synthesize and store triglycerides during periods of

food uptake and then, when food is not being absorbed from the intestinal tract, to release

fatty acids and glycerol into the blood so that they may be taken-up and used as substrates

for the harvest of energy (in the form of ATP). Here, this review will very briefly focus

on the two most important pathways involving fatty acids: fat synthesis (the formation of

triglycerides and incorporation into adipocytes) and fat catabolism (breakdown of fat

molecules in order to derive energy).

Fat synthesis (the production of triglyceride molecules) and integration into

adipocytes are now understood to be complex biochemical processes, with various

cellular and molecular players (Lefterova & Lazar, 2009). That being said, the pathway

through which fat synthesis occurs remains the same. The enzymes that mediate fat

synthesis are largely localized to the cytoplasm, whereas the breakdown of fat (as will be

discussed shortly) largely occurs because of enzymes in the mitochondria. Fatty acid

synthesis begins with cytoplasmic acetyl coenzyme A, which transfers its acetyl group to

another molecule of acetyl coenzyme A. This forms a four-carbon chain. Repetition of

this process builds up long-chain fatty acids (two carbons at a time). These newly

synthesized fatty acids are then transported across the membrane of adipocytes, where

they are combined with glycerol in an esterification reaction to form triglycerides, which

are then incorporated into these cells. The mechanism by which this occurs remains

unclear, with various proteins in the adipocyte plasma membrane being implicated in

fatty acid transport (Pohl, Ring, Korkmaz, Ehehalt & Stremmel, 2005).

Page 18: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

8

The utilization of fatty acids is a major source of energy in mammalian

physiology (Floyd, Medes & Weinhouse, 1947). Release of fatty acids from adipocytes is

the first major step in fat metabolism. Once the fatty acids have been taken-up by a

metabolic tissue (such as skeletal muscle or liver), they are transported into the

mitochondria, where ATP harvest occurs. The breakdown of a fatty acid is initiated by

linking a molecule of coenzyme A to the carboxyl end of the fatty acid. The coenzyme A

derivative of the fatty acid is subsequently taken through a series of reactions that are

collectively known as β-oxidation. In this process, a molecule of acetyl coenzyme A is

removed from the end of the fatty acid, which includes the transfer of two pairs of

hydrogen atoms. The hydrogen atoms from the coenzymes then enter the oxidative

phosphorylation pathway to form ATP. When an acetyl coenzyme A is split from the end

of a fatty acid, another coenzyme A is added, and the sequence is repeated. In this way,

the initial fatty acid is shortened by two carbons each time, until all the carbon atoms

have been transferred to coenzyme molecules. These molecules subsequently produce

carbon dioxide and ATP via the Krebs cycle and oxidative phosphorylation. The result is

a relatively large yield of ATP from a single fatty acid molecule, making β-oxidation of

fatty acids one of the most efficient methods of energy generation.

1.2.3. Bone

The final major component of body composition is bone, which makes up the

mammalian skeleton. Bone is a tissue that consists of a protein (collagen) matrix upon

which calcium salts (notably calcium phosphates) are deposited. For descriptive purposes,

a growing long bone is divided into the ends (epiphyses) and the remainder (the shaft).

Page 19: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

9

Bone remodeling is a complex process that mediates growth and loss of bone tissue

(Luisetto & Camozzi, 2009). Osteoblasts, the bone-forming cells at the shaft edge of the

epiphyseal growth plate, function by converting the cartilaginous tissues at the edge into

bone. Meanwhile, specialized cells called chondrocytes lay down new cartilage in the

interior of the plate. In this way, the epiphyseal growth plate remains intact, and is

gradually pushed away from the center of the bony shaft as lengthening of the shaft

occurs. Conversely, specialized cells called osteoclasts mediate a process called

resorption (Hattner, Epker & Frost, 1965). Here, osteoclasts function by removing the

mineralized matrix and breaking up organic bone. The minerals released during bone

resorption are subsequently transported to the blood. Bone growth and decline are

important indicators of overall health. Work in endocrinology has shown that bone

density and strength are strongly influenced by a variety of different hormonal

contributors (Olney, 2009). In this way, bone structure is a commonly used indicator of

endocrine status and possible pathology.

In summary, therefore, it can be seen that body composition is characterized by

different components, each with different properties. In the context of energy

homeostasis, most work is focused on delineating differences in skeletal muscle and

adipose tissue. White adipose tissue (WAT) serves as one of the most important stores of

energy. Skeletal muscle performs similar storage functions (although not to the degree of

adipose or liver), but is also capable of energy catabolism, and is one of the most

energetically demanding tissues in the body. In this way, constant interplay between

skeletal muscle and adipose has become a focus of metabolic research.

Page 20: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

10

1.3. Energy Balance and Metabolism

Animal physiology is predicated on the idea of homeostasis: the need for the body

to maintain a particular state, with perturbation resulting in deviation back towards the

mean or “steady state”. A delicate balance also maintains the state of energy within an

organism. The execution of various processes (both voluntary and involuntary) requires

energy (typically defined as ATP, but also in other forms). While the body expends

energy in order to conduct these processes, energy must also be taken in, in order to

replace that which is spent. This homeostatic mechanism is termed “energy balance”, and

maintenance of energy balance is under strict regulatory control within the organism.

Inability to maintain this balance can have disastrous consequences for overall health of

the organism (Spiegelman & Flier, 2001).

1.3.1. What is Energy Balance?

Animals intake energy sources (food) for the purposes of deriving various organic

molecules for use as fuel. The breakdown of these organic molecules (metabolism)

liberates the energy locked in their molecular bonds. This released energy is what cells

use in order to perform the various forms of biological work – including muscle

contraction, active transport within a cell, and molecular synthesis. Energy derivation and

expenditure within an organism is governed by the laws of thermodynamics, which

account for all energy in the universe. The first law of thermodynamics states that energy

can neither be created nor destroyed, but can only be converted from one form to another.

Therefore, the internal energy liberated during breakdown of an organic molecule can

either dissipate as heat, or be used to perform work (Spiegelman & Flier, 2001).

Page 21: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

11

The majority of energy derived from the breakdown of organic molecules is

utilized to perform work. The energy to be used for work must first be incorporated into

molecules of ATP. The subsequent breakdown of ATP serves as the immediate energy

source for the work. Biological work can generally be described as either “external” work

(ex. movement of limbs by contracting skeletal muscles); or “internal” work (cellular

activity, such as molecular transport and synthesis).

Thus, we can think of energy balance as a constant struggle between energy intake

and energy expenditure (Spiegelman & Flier, 2001). Energy intake is characterized by our

feeding habits, the nutrient sources that makeup (or are absent from) our diets.

Conversely, energy expenditure is the utilization of these nutrient sources for resting

metabolic processes, physical activity, and adaptive thermogenesis. “Tipping the scales”

too far in either direction can be thought of as maladaptive. If energy intake surpasses

energy expenditure, energy stores become larger and larger. In mammals, this can

manifest itself in metabolic syndrome, characterized by insulin resistance (and

subsequently diabetes) as well as obesity (significant fat accumulation). Similarly, if

expenditure of energy surpasses levels of intake, energy stores become depleted

(starvation). Without enough energy to maintain itself, a cell will eventually die.

Subsequently, if exacerbated, this would ultimately lead to coma and death of the

organism. Therefore, like all physiological systems under homeostatic control, energy

balance must be tightly regulated by complex mechanisms.

Page 22: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

12

1.3.2. Energy Intake

While human beings tend to think of their feeding behaviour as largely being

under conscious and voluntary control, the truth is that there are significant molecular

regulators of this process. Understanding these processes provides researchers with

significant insight about how the body manages to control the intake of food, and how

this can be affected by an individual’s energy expenditure. The molecular study of

feeding behaviour is made complex by the fact that this process is not only influenced by

unconscious, homeostatic direction, but also by psychological factors. In addition to food

availability, feeding is affected by metabolic, neuronal and hormonal factors; and is also

modified by powerful sensory, emotional and cognitive inputs. It is the interplay of all of

these factors (in higher-level organisms) that contributes to energy intake (Schwartz et al.,

2000).

In understanding the idea of physiological homeostasis, possibly no brain area has

received as much attention as the hypothalamus. This region of the brain is critical for

homeostatic processes such as feeding, thermoregulation, and reproduction (Elmquist et

al., 1999). The hypothalamus receives inputs from neural, endocrine and metabolic

signals. It then integrates this information and triggers various response pathways. The

role of the hypothalamus in regulating feeding behaviour is well established. Lesion

studies demonstrate that lesions in the ventromedial hypothalamus result in obesity, while

lesions of the lateral hypothalamus cause leanness (Elmquist et al., 1999). We now

understand that the hypothalamus is able to regulate feeding through secretion of various

neuropeptides. These neuropeptides are essentially neurotransmitters, which function

through complex metabolic signaling pathways. They are classified as either being

Page 23: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

13

orexigenic (appetite-stimulating) or anorexigenic (appetite-inhibiting). The most

prominent hypothalamic orexigenic neuropeptide is neuropeptide Y (NPY). When

administered into cerebral hemispheres or specific hypothalamic nuclei, NPY robustly

and rapidly increases feeding and suppresses energy expenditure, thereby promoting

obesity (Stanley et al., 1986; Billington et al., 1994). Conversely, the major anorexigenic

neuropeptides produced by the hypothalamus are cocaine and amphetamine related

transcript (CART), and α-melanocyte stimulating hormone (α-MSH), which are both

derived from proopiomelanocortin (POMC). These neuropeptides function by activation

of melanocortin 4 receptor (MC4R). Activation of MC4R by α-MSH reduces food intake,

while suppression of MC4R by pharmacological antagonists increases feeding (Fan et al.,

1997). Similarly, genetic deletion of MC4R results in obesity in mice (Huszar et al.,

1997), while this same phenotype also occurs due to mutation in the POMC gene,

disrupting synthesis of CART/α-MSH (Krude et al., 1998). Thus, the hypothalamus is

capable of regulating feeding through secretion of neuropeptides, which then have

downstream effects upon metabolic signaling pathways.

But how does the hypothalamus know whether to secrete orexigenic or

anorexigenic neuropeptides? It is this question that is critical to understanding the

homeostatic control of energy balance. The hypothalamus secretes neuropeptides in

response to external molecular signals. The most prominent of these signals are endocrine

in nature. It was the discovery of the adipose-derived hormone leptin that provided the

best evidence for endocrine control of feeding (Friedman & Halaas, 1998). Leptin is

primarily manufactured in the adipocytes of WAT, and therefore the level of circulating

leptin is directly proportional to the total amount of fat. With regards to feeding

Page 24: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

14

behaviour, leptin serves as a satiety signal, inhibiting appetite and further food intake.

Mice with homozygous mutations in the Ob gene, which codes for the leptin protein,

develop obesity characterized by hyperphagia (Zhang et al., 1994). Treatment of these

mutant mice with the missing hormone attenuates weight gain and appetite (Halaas et al.,

1995). Expression of leptin is found in other tissues, such as skeletal muscle, suggesting

that other metabolic organs may act to inhibit appetite (Wang et al., 1998). The

mechanism through which leptin suppresses appetite is not entirely clear. However, what

is known is that leptin binds to receptors in the hypothalamus, and regulates secretion of

hypothalamic neuropeptides. For example, leptin administration has been shown to

reduce expression of orexigenic neuropeptides, such as NPY (Elias et al., 1998).

Conversely, leptin action on the hypothalamus increases expression of anorexigenic

peptides, such as CART and α-MSH (Elmquist et al., 1999). Through these simultaneous

effects, leptin is able to reduce food intake through appetite suppression.

In contrast to leptin is the hormone ghrelin. This peptide hormone is expressed

primarily in stomach and brain (with the stomach being its major site of synthesis), but

the control of its expression is not completely understood (Lutter et al., 2008). Ghrelin

action opposes that of leptin, acting on the hypothalamus to induce hyperphagia and

energy intake (Nakazato et al., 2001). Daily administration of ghrelin causes increased

food intake, reduced fat utilization, and eventual obesity in mice and rats (Tshcop, Smiley

& Heiman, 2000). Similar to leptin, however, the mechanism of ghrelin action has not

been completely elucidated. There is existing evidence to suggest that ghrelin acts upon

the hypothalamus to increase NPY release, and stimulation of hunger (Tschop et al.,

Page 25: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

15

2000). Thus, it can be concluded that energy intake is largely controlled by neuronal and

endocrine signals, with the overall purpose being the maintenance of energy homeostasis.

1.3.3. Energy Expenditure

In order for the organism to maintain energy homeostasis, the energy generated

through intake (i.e. feeding) must be split in order to drive various processes. Energy

expenditure is largely accomplished by three major factors: basal metabolism, physical

activity, and adaptive thermogenesis. Here we will briefly review each of these factors,

and how they can contribute to overall energy balance. When quantifying the energy of

metabolism, scientists will often use a particular unit of measurement, referred to as the

calorie. Energetically speaking, one calorie is the amount of heat required to raise the

temperature of one liter of water by one degree Celsius. The energy stores in food are

quite high relative to a calorie, so in the context of energy balance, the term kilocalorie

(Calorie) will be used. Total energy expenditure is often described in Calories, with

expenditure per unit of time referred to as the metabolic rate.

Many factors influence overall metabolic rate, including age, sex, height, and

overall health (Spiegelman & Flier, 2001). Thus the most common methodology for

evaluating it specifies certain standardized conditions and measures what is known as the

basal metabolic rate (BMR). This is often termed the ‘metabolic cost of living’ and refers

to the energy necessary to sustain autonomic cellular processes in major energetically

demanding tissues such as heart, liver, kidney and brain. When in the basal condition, the

subject is at mental and physical rest in a room at a comfortable temperature, and is often

in the fasted state (i.e. having not eaten for at least 12 hours). When animals are digesting

Page 26: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

16

food, but not expending energy through other means (such as physical activity), energy

expenditure is defined as ‘resting metabolic rate’ (RMR). Several factors are known to

influence BMR (Spiegelman & Flier, 2001). The thyroid hormones are the single most

important determinant of BMR, regardless of an organism’s size, age, or sex.

Hyperthyroidism (pathological secretion of thyroid hormone) increases oxygen

consumption and heat production, with the overall result of net cellular catabolism and

loss of body weight (Kahaly, 2010). Another major regulator of BMR is the

hormone/neurotransmitter epinephrine. Release of epinephrine occurs during sympathetic

stimulation of the autonomic nervous system, resulting in increased heart rate, contraction

of blood vessels, and dilation of air passages. Epinephrine stimulates glycogen and

triglyceride catabolism, since ATP splitting and energy liberation occur during

breakdown and re-synthesis of these metabolic substrates. Thus, we can see that BMR is

determined through the interaction of several influencing factors, and can control an

organism’s basic level of energy expenditure.

‘Non-resting’ conditions are those that occur during periods of activity. The factor

that can most increase metabolic rate (energy expenditure) is altered skeletal muscle

activity. Even minimal increases in muscle contraction can significantly increase

metabolic rate, and strenuous exercise might raise energy expenditure more than

fifteenfold (Jones & Killian, 2000). While we have already described metabolic

differences in fiber-type, and will later discuss the metabolic potential of muscle, it is

important to know how physical activity (particularly exercise) can influence energy

homeostasis, and the role of other important organs in mediating these effects. During

exercise, large quantities of fuels must be mobilized to provide required energy for

Page 27: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

17

muscle contraction. These fuels include circulating plasma glucose, fatty acids, and

glycogen (stored glucose polymers). A major role is played by the liver in regulating this

process (Jones & Killian, 2000). The plasma glucose used during exercise is largely

provided by the liver, both by breakdown of its glycogen stores, and by gluconeogenesis

(synthesis of new glucose molecules). Lipolysis of triglycerides creates a surplus of

glycerol for the liver, and fatty acids for metabolism by muscle. Changes in plasma

glucose during exercise are small in the short-term, but long-term exercise does lead to

transient decreases in blood glucose, due to an inability of hepatic gluconeogenesis to

keep up with glucose utilization. The metabolic profile seen in an exercising person

(increased hepatic gluconeogenesis, triglyceride breakdown, fatty acid utilization, etc) is

similar to that seen in a fasting person. This relates back to the main equation of energy

balance. An energy deficit (and its resultant metabolic profile) can be achieved through

increased energy expenditure, or decreased energy intake.

In earlier discussion on thermodynamics, it was mentioned that liberated energy

could be either used for work or heat. It has been demonstrated that liberated energy from

the breakdown of organic molecules can be used to create ATP (which are split to

perform work). However, as thermodynamics tells us, energy can also be lost in the form

of heat. In the context of metabolism, this heat loss leads to discussion of the third and

final component of energy expenditure: adaptive thermogenesis. Physiologically,

thermogenesis is primarily controlled by brown adipose tissue (BAT). The evolutionary

importance of BAT and adaptive thermogenesis is clear: it functions to generate body

heat, and is particularly abundant in newborns and hibernating animals, which are

incapable of shivering (Enerback, 2010). Mitochondrial density is significantly greater in

Page 28: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

18

BAT than WAT. It is through mitochondria that BAT is able to generate heat.

Mitochondria are the major energy generators in the cell, and function to produce ATP

through glycolysis and fatty acid oxidation. This is accomplished by movement of

protons across a gradient and into the mitochondrial matrix. However, protons may “leak”

back across the inner mitochondrial membrane in a manner not linked to ATP production.

This uncouples energy storage from oxygen consumption, a process referred to as

‘uncoupled respiration’. Inherently, a certain degree of uncoupled respiration is expected

to occur in mitochondria of all cells. However, this process is greatly accelerated in BAT,

due to high expression of uncoupling proteins (UCP), which function as specialized

protein channels that provide a route for protons to return across the mitochondrial

membrane. The end result is energy expenditure, but largely in the form of heat. Mice that

lack BAT are prone to obesity, suggesting that this mechanism is an important avenue of

energy expenditure (Lowell & Flier, 1997). Evidence now exists for several different

types of UCP. UCP1 is predominantly found in BAT, while UCP2 is expressed in a

variety of different tissues (Enerback et al., 1997). UCP3 is the major form found in

skeletal muscle, and muscle-specific overexpression of UCP3 results in leanness and

hyperphagia (Clapham et al., 2000). Thus, it seems as though thermogenesis occurs in

tissues other than BAT, and elucidating these processes will provide scientists with a

greater understanding of whole-body energy expenditure.

1.3.4. Energy Balance, Body Composition and Obesity

Understanding the key molecular players governing whole-body energy intake

and energy expenditure are key for translational study of body composition. Deregulation

Page 29: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

19

of energy balance results in manifestation of changes in body composition, and

subsequently impacts overall health (Friedman, 2009). As mentioned, when energy

expenditure exceeds energy intake, the result is cell death due to insufficient energy. If

persistent, death of the organism will occur. However, the more pressing problem (at least

in Western society) is a deregulation in energy balance caused by an increase in energy

intake, and subsequently limited energy expenditure. The result is increased adiposity (fat

accumulation), higher levels of circulating glucose, and increased glycogen storage. Over

time, this results in obesity, type 2 diabetes mellitus (T2DM), and metabolic dysfunction.

Obesity (excess fat accumulation) is a major health concern. Those who suffer

from obesity are at significantly greater risk to develop heart disease, cancer, and T2DM.

The prevalence of obesity has increased steadily, particularly in developed, Western

countries (Friedman, 2009). While the etiology of obesity is complex (linking both

genetic susceptibility and environmental factors), at its root, all instances of obesity are

characterized by a dysfunction in energy balance. A chronic imbalance between energy

intake and energy expenditure can lead to an increase in both fat cell size and fat cell

number. Therefore, a successful obesity therapy must impact energy intake, energy

expenditure, or both.

The use of drugs that affect energy intake have been met with limited success.

After the isolation of leptin came the promise that exogenous administration of this

hormone might be capable of inhibiting appetite and inducing weight loss (Friedman &

Halaas, 1998). However, long-term treatment of subjects with leptin results in reduced

efficacy, due to the development of leptin resistance (Mantzoros & Flier, 2000). Scientists

believe that leptin resistance (due to higher levels of the circulating hormone from

Page 30: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

20

increased adipose tissue) may exacerbate energy intake in obese individuals. Thus,

scientists have instead focused upon the discovery of leptin receptor agonists. This

avenue has yielded some surprising insights, as some cytokines have been shown to have

leptin-like effects, suppressing appetite and promoting weight loss. Most notable among

these is ciliary neurotrophic factor (CNTF), a neurocytokine that has been shown to

reverse obesity in leptin-resistant mice (Gloaguen et al., 1997), although the mechanism

of action of these cytokines is unknown. With that in mind, some scientists have

suggested that a more appropriate method might be to simply target hypothalamic

neuropeptides themselves, as the secretion of these neuropeptides is what is controlled by

the endocrine signals. To that end, there has been development of small-molecule

antagonists for NPY, the major orexigenic neuropeptide; as well as agonists for the

MC4R receptor, which has anorexigenic effects on appetite (Gehlert, 1999). Only time

will tell whether success in regulation of energy balance can be efficiently achieved

through modulation of feeding behaviour.

On the other hand, some believe that treatments geared toward increasing

metabolic rate (and energy expenditure) may be a more promising avenue for the

treatment of metabolic disease. Feeding behaviour is still influenced by social and

environmental factors, but this is less applicable for regulation of metabolic rate. As body

weight declines, metabolic rate subsequently decreases as well, and therefore a strong

treatment may be used to either prevent this drop, or increase overall metabolic rate.

Although thyroid hormone is the main regulator of BMR, use of exogenous (i.e.

supraphysiological) doses of thyroid hormone can have negative side-effects, including

goiter, loss of lean muscle mass, and blindness (Kahaly, 2010). Conversely, physicians

Page 31: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

21

often recommend that patients who suffer from hypometabolism take part in resistance

exercise training regimens, with the purpose of building lean muscle (Harrison &

Leinwand, 2008). This is because skeletal muscle is a major metabolically demanding

tissue, and increased muscle mass is sufficient to induce significant changes in resting

metabolism. Later discussion in this review will focus on how skeletal muscle can

influence systemic metabolism, but for now it is worth mentioning that scientists have

begun to focus on manipulation of this tissue as a possible method of regulating energy

expenditure and overall energy homeostasis. Factors that influence skeletal muscle

growth (such as androgens) may be looked upon as treatment measures. By targeting the

energy balance equation, scientists are attempting to find novel methodologies for treating

metabolic disease.

1.4. The Androgen System

The term ‘androgens’ is used to collectively refer to the class of male sex

hormones. Most prominent of the androgens are the hormones testosterone (T) and

dihydrotestosterone (DHT), which are known to have diverse functions in mammalian

physiology. These can be contrasted with the estrogens, the class of female sex hormones

(including 17β-estradiol and estrone). Much is known about androgens, and a large

amount of work has elucidated details regarding their pharmacology, synthesis and

mechanism of action.

Page 32: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

22

1.4.1. Androgen Pharmacology – Structure and Production

Androgens fall into a class of hormones termed ‘steroid hormones’. Included in

this class are not only the male sex hormones, but also the female sex hormones,

glucocorticoids and mineralocorticoids. All hormones within this class have certain

characteristics with regards to their structure and stability (McEwen, 1992). Steroids are

lipid molecules, and all of these hormones are composed of four interconnected rings of

carbon atoms. A few polar hydroxyl groups may be attached to these rings, however the

overall number of hydroxyl groups are not numerous enough to make steroids into polar

molecules. For this reason, sex hormones are not soluble in the blood, but rather need to

be transported by binding to an albumin (water-soluble protein), known as sex hormone-

binding globulin (SHBG). However, the lipid, non-polar nature of steroid hormones

allows them to diffuse easily across the cell membrane, unlike peptide hormones (a

separate class that includes insulin and glucagon).

Steroidogenesis describes the biochemical process by which steroids are generated

from cholesterol, and subsequently transformed into other steroids (Aizawa et al., 2008).

There is no specific gene that codes for androgen, but rather androgens are derived by

enzymatic reactions from lipid precursors. In the case of de novo androgen synthesis (in

males), this process takes place in the leydig cells of the testes and (to a significantly

lesser degree) in the adrenal cortex. Cholesterol precursors are initially cleaved by

CYP11A, a mitochondrial cytochrome P450 oxidase. The loss of 6 carbon atoms from the

cholesterol molecule results in the prohormone pregnenolone. The cleavage of two more

carbons from this precursor by the CYP17A enzyme yields a variety of 19-carbon

steroids. Subsequent oxidation of the 3-hydroxyl group by the enzyme 3-β-

Page 33: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

23

hydroxysteroid dehydrogenase produces androstenedione. Finally, in the rate-limiting

step, this intermediate is reduced by 17-β-hydroxysteroid dehydrogenase to yield T. Thus,

T is not produced directly by protein synthesis from a gene, but rather it is the expression

of these important enzymes that regulate its biosynthesis.

What is important to remember is that steroid hormone biosynthesis does not end

with T production. T itself, despite having its own functions, also acts as a precursor to

other sex hormones. In fact, T serves as the major precursor for the synthesis of estrogens.

This reaction is mediated by the enzyme aromatase, which oxidizes the T molecule and

removes a methyl group, resulting in the production of estradiol. Female aromatase-null

(ArKO) mice demonstrate significantly elevated T levels, significantly reduced estrogen

levels, and severe defects in sexual differentiation, such as underdeveloped external

genetalia and uteri (Fisher, Graves, Parlow & Simpson, 1998). Expression of aromatase is

highest in the gonads, but it is also found in brain, adipose tissue, placenta and

endometrium.

Another major metabolite of T is the androgen dihydrotestosterone (DHT), which

is largely expressed in prostate gland, testes, hair follicles, and adrenal glands (Zitzmann,

2009). Conversion of T to DHT occurs via the enzyme 5-α-reductase, which reduces the

4,5 double-bond in the T molecule through the addition of two hydrogen atoms. DHT

functions in a similar fashion as other androgens, but differs from T in that it is

considered to be more potent. This is for two primary reasons. First, DHT has stronger

affinity for androgen receptor (AR), which is the nuclear receptor that mediates androgen

action; as well as for SHBG, which is the major transporter of androgens through the

bloodstream. Secondly, unlike T, DHT cannot be aromatized to estrogens, and thus does

Page 34: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

24

not serve as a precursor, which would limit its yield. DHT’s increased potency has made

it a target for treatment of disorders associated with hyperandrogenicity, such as benign

prostatic hyperplasia (prostate growth) and androgenic alopecia (male-pattern baldness).

This is typically accomplished through the use of a 5-α-reductase inhibitor, which inhibits

the enzyme, and subsequently reduces conversion of T to DHT (Walsh, 2010).

Secretion of androgens from the testes is under strict homeostatic control in

normal males (Tobet, Bless & Schwarting, 2001). Similar to release of glucocorticoids

from the adrenal glands, androgen secretion is controlled by hormonal signals from the

brain (specifically the hypothalamus and the pituitary gland). Together, these hormones

constitute a feedback loop that allows them to regulate one another. Gonadotropin-

releasing hormone (GnRH) is released from hypothalamic nuclei, which then travels to

the anterior pituitary gland. Stimulation of the anterior pituitary by GnRH results in the

release of the gonadotropins: luteinizing hormone (LH) and follicle-stimulating hormone

(FSH). As the name suggests, the gonadotropins stimulate the gonads. In males, LH acts

upon the leydig cells of the testes, and contributes to the production of T and other

androgens. In contrast, FSH stimulates maturation of the seminiferous tubules of the

testes in males, and also acts upon sertoli cells, functioning to promote spermatogenesis.

LH also indirectly plays a role in spermatogenesis, as it stimulates T production, and

androgens are involved in regulating this process. Also, T acts upon hypothalamic AR in

order to reduce secretion of GnRH. This negative feedback maintains concentrations of

these hormones in homeostatic balance, thus allowing for fulfillment of normal

androgenic function.

Page 35: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

25

1.4.2. Androgen Receptor

As mentioned, androgens exert their functions by binding to the androgen receptor

(AR). Like other sex steroid hormone receptors, AR is a ligand-inducible transcription

factor belonging to the nuclear receptor subfamily (Chang, Kokontis & Liao, 1988). The

gene encoding AR is located at position q11-12 of the X chromosome, and is comprised

of eight exons. It encodes a protein composed of 919 amino acids, with a molecular mass

of approximately 110 kDa (Lubahn et al., 1988).

The structure of the AR protein itself is not too dissimilar from that of other

nuclear receptors (Chawla, Repa, Evans & Mangelsdorf, 2001). It consists of four

functional domains: an amino-terminal transactivation domain (NTD, encoded by exon

1), containing stretches of glutamine, proline and glycine; a highly conserved central

DNA-binding domain (DBD, encoded by exons 2 and 3); a hinge region (encoded by the

5’ part of exon 4); and a carboxy-terminal ligand-binding domain (LBD, encoded by the

3’ part of exon 4, as well as exons 5-8), to which the androgen binds.

Androgens (such as T and DHT) are lipid-based molecules, and as such, can

easily diffuse across the lipid-bilayer membrane of the cell (without the aid of a transport

protein). Upon binding of the androgen to the LBD of AR, an important sequence of

events is initiated (Lee & Chang, 2003). When not bound to its androgen ligand, AR is

maintained in a stable conformation in the cytoplasm through association with chaperone

molecules. The specific chaperone molecule involved is usually dependent upon the cell

type (for example, heat shock protein 70 maintains AR conformation in prostate cells –

see He et al., 2004). Upon binding of the androgen ligand, the conformation of AR

undergoes change and the chaperone molecule disassociates (Chang et al., 1995). AR

Page 36: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

26

then forms a homodimer, which translocates to the nucleus. This homodimer then binds

to specific DNA motifs, termed androgen response elements (AREs), which then results

in recruitment of various coactivators to form the transcription complex (Roche, Hoare &

Parker, 1992). These AREs are situated in or near various target genes, and largely fall

into two classes. The ‘classical’ AREs are also recognized by other steroid receptors

(Claessens et al., 2001), while ‘selective’ AREs are only responsive to activation by AR

(Shaffer et al., 2004).

Expression of AR is found in many tissues of the body, albeit to different degrees

(Ting & Chang, 2008). The highest expression of AR is believed to be found in the

adrenal gland, followed by epididymis, prostate, skeletal muscle, kidney, liver, and heart.

With respect to skeletal muscle, higher expression of AR is found in androgen-dependent

muscles (such as the levator ani) as compared to non-androgen-dependent muscles, such

as the extensor digitorum longus (EDL) (Monks, Kopachik, Breedlove & Jordan, 2006).

AR expression is also controlled by androgens themselves. Androgens have been shown

to regulate both AR mRNA as well as protein. However, it seems as though the effects

that androgens have on AR expression (i.e. whether they increase it or decrease it) are

tissue-dependent. Treating human prostate cancer cells with T reduces AR mRNA almost

two-fold (Trapman et al., 1990). Conversely, androgen action increases AR expression in

human hepatocellular carcinoma cells (Wiren et al., 1997), as well as in rat hippocampus

(Kerr, Allore, Beck & Handa, 1995). In skeletal muscle, it appears that both T and DHT

increase AR mRNA expression (Lee & Chang, 2003).

The binding of the homodimer androgen-AR complex with AREs (and subsequent

modulation of gene expression) characterizes the most well-understood mechanism of AR

Page 37: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

27

function. However, recent evidence has uncovered non-transcription related (termed

‘non-genomic’) functions of AR (Baron et al., 2004). These non-genomic mechanisms

typically involve interactions of AR with other cell-signaling pathways. This includes

increases in intracellular calcium, and activation of various kinase enzymes, such as

protein kinase A, protein kinase C, and MAP kinase; leading to diverse cellular effects,

such as smooth muscle relaxation, neuromuscular and junctional signal transmission, and

neural plasticity (Heinlein & Chang, 2002). These T-dependent effects on second

messenger signals are unaffected by inhibitors of transcription and translation, and occur

within seconds to minutes, which are time courses that are believed to be too rapid to

represent changes in gene expression (Fix, Jordan, Cano & Walker, 2004). Thus it

appears that AR does not simply exert itself through modulation of gene expression

(although this does represent its major function), but also plays a role in regulation of cell

signaling.

1.5. Androgens, AR and Muscle

As mentioned previously, androgens have notable effects on body composition.

Perhaps most well-studied of these effects is the anabolic actions of androgens on muscle.

Muscle has long been known to be a prominent androgen target. AR expression has been

demonstrated in all three types of muscle: skeletal, cardiac and smooth (Lee & Chang,

2003). The functions of AR in these types of muscle, and the subcellular mechanisms

involved, are amazingly diverse.

Page 38: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

28

1.5.1. Anabolic Functions of AR in Muscle

The classical role of muscle AR is its anabolic function. It has largely been

demonstrated that androgen/AR action is responsible for development and maintenance of

muscle fibers (Bhasin et al., 1997; MacLean & Handelsman, 2009). There is considerable

evidence suggesting that androgens promote not only myogenesis by acting upon

progenitor satellite cells, but also mediate hypertrophy of mature individual muscle fibers.

While muscle growth during mammalian development is largely due to myogenesis and

the creation of new muscle fibers, growth in adulthood is largely believed to be due to

hypertrophy of existing fibers (Sinha-Hikim et al., 2002). Various molecular and cellular

mechanisms have been implicated in mediating each of these effects, in addition to

overlapping mechanisms that control both myogenesis and fiber hypertrophy.

Singh et al. (2003) showed that T has the capability to promote differentiation of

mesenchymal pluripotent stem cells toward the myogenic lineage, thereby driving

myogenesis. Further research has shown that similar effects can be found with regards to

development of satellite cells, when treated with T. Satellite cells are found between the

basal lamina and the plasma membrane of muscle fibers (Mauro, 1961). These specialized

progenitor cells have the ability to differentiate into muscle fibers, or fuse and augment

existing fibers. In this way, they can be likened to adult stem cells, and represent the

major source for the addition of new myonuclei into the growing muscle fiber. Satellite

cells are maintained in a quiescent state, until activated by external signals, which then

promote myogenesis (Tedesco, Dellavalle, Diaz-Manera, Messina & Cossu, 2010).

There are several lines of evidence that have been used to show that androgens

can act as an external signal to stimulate the proliferation of satellite cells in muscle. First,

Page 39: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

29

AR expression has been readily demonstrated in satellite cell cultures, suggesting that

these precursor cells do serve as androgen targets (Doumit, Cook & Merkel, 1996). Work

with cultured human satellite cells has shown that AR protein expression increases when

cells are exposed to T (Sinha-Hikim, Taylor, Gonzalez-Cadavid, Zheng & Bhasin, 2004).

Powers and Florini (1975) were the first to successfully demonstrate that T, but not

estradiol, was capable of stimulating the mitotic activity of satellite cells in established

myoblast culture systems. Similarly, Sinha-Hikim and colleagues (2003) showed that, in

men, T supplementation led to a significant increase in the total number of satellite cells

found. Thus, effects of T on myogenesis (and proliferation of myogenic factors) have

been well established.

Whether androgens are capable of regulating hypertrophy of adult myofibers is

less clear. Most of the work in this context has been performed on human populations,

and have involved exogenous T-treatment, making it difficult to tease out effects on

myogenesis from those on muscle hypertrophy. Some evidence indicates that T induces

the hypertrophy of both type I (oxidative) and type II (glycolytic) fibers. When comparing

muscle biopsies from athletes who abuse anabolic steroids against those who have not

used such supplementation, the greatest disparities are seen in type II muscle fibers (Kadi,

2008). However, this belief is rather controversial, and there is some evidence suggesting

that androgens may have their primary effects on type I fibers. In comparison of biopsies

taken from trapezius muscle of androgen users and controls, the largest disparity is seen

between type I fibers, although there is also a significant difference in size of type II

fibers (Kadi, Eriksson, Holmner & Thornell, 1999). Similar differences are also seen in

the vastus lateralis muscle (Eriksson, Kadi, Malm & Thornell, 2005). It is believed that

Page 40: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

30

this more robust effect on type I fibers is due to the fact that these fibers are more

sensitive to the effects of T treatment (Hartgens & Kuipers, 2004). For example, Sinha-

Hikim and colleagues (2002) found that type II fibers required twice as much exogenous

T as type I fibers in order to demonstrate hypertrophy. However, the above study also

found no differences in fiber type proportion, a finding that is disputed by related studies

(Kadi et al., 1999). Thus, specific effects of androgens upon muscle fiber type (both in

terms of proportion of total fibers and hypertrophy of individual fibers) remain unclear.

Further evidence indicates that activation of AR is necessary for promotion of

androgen’s anabolic functions upon skeletal muscle. Blockade of AR through the use of

the AR-antagonist oxendolone suppresses muscle hypertrophy of rat muscle fibers (Inoue,

Yamasaki, Fushiki, Okada & Sugimoto, 1994). Also, knockout of AR (either

ubiquitously, or selectively in myocytes) results in muscular atrophy and reduced skeletal

muscle strength (MacLean et al., 2008; Ophoff et al., 2009). Similar to precursor satellite

cells, AR expression can be modulated by T administration, as mentioned previously.

AR-containing myonuclei in human skeletal muscle become more numerous after the use

of anabolic steroids (Kadi, Bonnerud, Eriksson & Thornell, 2000). These results were

consistent among biopsies taken from the neck (trapezius muscle), as well as the limb

(vastus lateralis).

These anabolic effects are seemingly not limited to skeletal muscle, and androgens

have also been shown to be important in promoting hypertrophy of cardiac muscle. In

vitro work performed on cultured human cardiomyocytes found that T administration

resulted in increased incorporation of [3H]phenylalanine into cardiomyocytes, as well as

atrial natriurectic peptide secretion – both of which are significant indicators of

Page 41: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

31

cardiomyocyte hypertrophy (Marsh et al., 1998). These T-mediated effects were

subsequently abolished when the cultured cells were first treated with the anti-androgen

cyproterone acetate. These findings were then confirmed by in vivo work performed on

ARKO mice. Whole-body ARKO mice demonstrate impaired cardiac growth, and smaller

overall heart mass (Ikeda et al., 2005). In addition, various cardiac impairments are found

in these ARKO mice, including cardiac fibrosis and impaired ventricular function. Taken

together, these results indicate that androgen-AR signaling is necessary for normal

cardiomyocyte growth and heart function. However, it also suggests that

supraphysiological doses of T may result in pathological hypertrophy of cardiac muscle,

providing a potential explanation for the increased incidence of sudden cardiac death in

athletes that abuse anabolic steroids (Luczak & Leinwand, 2009). Relatively little

research has been conducted in this area, and any links between androgens and

hypertrophic cardiomyopathy (and associated heart disease) are surrounded by a large

degree of controversy.

1.5.2. Muscle AR and Neuromuscular Development

Androgens and AR are important regulators of sexual differentiation, and strongly

contribute to the development of sexually dimorphic muscles, as well as the motoneurons

that innervate them. Most well-studied of these muscle systems is the spinal nucleus of

the bulbocavernosus (SNB) neuromuscular system (Sengelaub and Forger, 2008). These

motoneurons project from the lumbar spinal cord into the striated muscles of the

perineum: the bulbocavernosus (BC) and levator ani (LA). The muscles themselves are

attached to the base of the penis, and their contraction mediates male sexual behaviour.

Page 42: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

32

SNB neurons are larger and more numerous in males than females (Breedlove & Arnold,

1980). In rats, during gestation, both sexes possess the SNB motoneurons and target

muscles, but prenatal T secretion allows for the neuromuscular system to be maintained in

males, whereas females undergo apoptosis (Nordeen, Nordeen, Sengelaub & Arnold,

1985). Not surprisingly, the development of the motoneurons that innervate the sexually

dimorphic muscles (BC/LA) are also under the control of circulating steroid hormones

(Breedlove & Arnold, 1980). These motoneurons innervate the striated BC and LA

muscles, and mediate copulatory behaviour in males. Conversely, in females, the SNB

motoneurons develop prenatally, but undergo significant postnatal cell death with

significantly fewer neurons seen in adult females, as compared to age-matched males

(Nordeen et al., 1985). Thus, in adulthood, SNB motoneurons are larger in number and

size in males (Breedlove & Arnold, 1980; McKenna & Nadelhaft, 1986).

It is now well-accepted that the survival of SNB motoneurons and their target

musculature occurs in males due to actions of circulating androgens in early postnatal

development (Sengelaub & Forger, 2008). Activation of AR by androgens is necessary

for masculinization of the SNB system to occur, and evidence for this is demonstrated

through the use of androgen-insensitive Testicular feminization mutation (Tfm) male rats.

The use of Tfm male rats has supported the necessary role of androgens/AR in

development and masculinization of the nervous system (Zuloaga, Puts, Jordan &

Breedlove, 2008). Tfm males, which have non-functional AR, demonstrate a feminized

SNB system, further highlighting the fact that this effect is AR-mediated (Breedlove &

Arnold, 1981). Similarly, administering the potent AR-antagonist flutamide to neonatal

male rats also suppresses SNB development (Breedlove & Arnold, 1983). However,

Page 43: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

33

despite the fact that activation of AR is a requisite of SNB development, it is not yet clear

which tissues or cells are necessary site(s) of androgen action.

Several lines of evidence indicate that AR in the perineal muscles themselves

(BC/LA) are the necessary site of androgen action. AR expression is significantly higher

in sexually dimorphic muscles (such as the LA), as compared to non-sexually dimorphic

muscles (such as the EDL), with enrichment at the neuromuscular junction (Monks,

O’Bryant & Jordan, 2004). Furthermore, AR immunoreactivity is found in perineal

muscles during the critical postnatal period for SNB development (Jordan, Padgett,

Hershey, Prins & Arnold, 1997). Removal of the BC/LA muscles on the first day of birth

results in a dramatic loss of SNB motoneurons by PND10, an effect that could not be

rescued by subsequent T treatment (Kurz, Cover & Sengelaub, 1992). Finally, injection of

flutamide (an AR antagonist) directly to the perineum of females results in fewer SNB

motoneurons than when flutamide is delivered systemically (Fishman & Breedlove,

1992). Taken together, these results suggest that AR in muscle fibers plays a critical role

in mediating development of the SNB neuromuscular system. However, in contrast to this

theory, Niel and colleagues (2009) found that transgenic expression of AR only in muscle

fibers of Tfm males was not capable of rescuing the SNB system in these mutants. Thus,

while significant evidence suggests the involvement of muscle AR in SNB development,

it appears that it is not sufficient, and AR in other tissue(s) may also be responsible for

this effect.

Page 44: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

34

1.5.3. Spinal and Bulbar Muscular Atrophy

Up to this point, we have shown that androgens/AR have significant effects upon

normal muscular development in males. Genetic (XY) males that harbor mutations in the

AR gene (or who are unable to synthesize androgens) demonstrate severe defects in these

contexts. However, androgens are also explored in the field of neuromuscular pathology,

as they have become linked with a disease known as Spinal and Bulbar Muscular Atrophy

(SBMA), also known as ‘Kennedy’s Disease’.

SBMA is a progressive neurodegenerative disease, largely affecting males, which

is characterized by progressive weakness and motoneuron death (Kennedy et al., 1968).

Prevalence of SBMA is very rare, with manifestation of the disease in only 1/400,000

(Fischbeck, 1997). Genetic analysis of SBMA first found that the disease was X-linked,

through localization of the defective allele using polymorphism linkage analysis

(Fischbeck et al., 1986).

It is now understood that the development of SBMA is related to the number of

glutamine (CAG) repeats in the AR gene (Monks et al., 2008). Polyglutamine repeats

exist in the previously discussed NTD region of the AR gene. The existence of this repeat

region is thought to contribute to differences between individuals in androgen sensitivity.

Shorter AR repeats increase receptor transactivation, with longer repeats decreasing

transactivation. After the determination of the X-linked inheritance pattern of SBMA,

scientists eventually discovered that the disease is associated with an increased number of

CAG repeats in exon 1 of the AR gene (La Spada et al., 1991). Not only that, but the

number of glutamine repeats in the gene was predictive of age of onset, as well as

severity of the disease, with an increasing number of CAG repeats indicative of earlier

Page 45: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

35

age of onset and increased severity (Atsuta et al., 2006; La Spada et al., 1992). Mouse

models that attempt to recapitulate this disease, through induction of glutamine repeats in

the AR gene, do show some of the symptoms of SBMA (Sopher et al., 2004).

The mechanism of how polyglutamine expansion in the AR gene subsequently

results in neurodegeneration and cellular death remains controversial. The prevailing idea

within the field is that the disease is neurogenic (Monks et al., 2008). That is, the

pathology manifests itself primarily in motoneurons, with the resultant toxicity causing

motoneuron death and subsequent muscular atrophy as a secondary result of this

motoneuron death (Monks et al., 2008). This is supported by in vitro work demonstrating

that polyglutamine expanded AR are sufficient to cause motoneuron death in cultured

motoneurons (Merry et al., 1998). However, recent work has cast doubt on this initial

premise. Most notably, polyglutamine expansion of the AR gene only in motoneurons of

transgenic mice is not capable of inducing symptoms of SBMA (Abel et al., 2001).

It now appears that SBMA may be myogenic in nature (i.e. it manifests itself

initially in muscle, with muscular atrophy resulting in motoneuron death). It has been

shown experimentally in an SBMA transgenic mouse model that muscular atrophy does

in fact precede motoneuron death (Yu et al., 2006). Furthermore, overexpression of AR

(without polyglutamine expansion) in muscle fibers creates a phenotype consistent with

most symptoms of SBMA (Monks et al., 2007). The resultant myopathy in this model is

associated with deregulation of gene expression in muscle fibers, and mitochondrial

abnormalities (Musa et al., In Prep). Thus, although SBMA appears to be AR-mediated,

the site-specific etiology of this disease remains unclear. Further characterization of

Page 46: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

36

disease models, as well as patient populations, will be necessary in order for scientists to

gain a better understanding of this neuropathology, and subsequently design treatments.

In summary therefore, androgen action on skeletal muscle is important for

mediating a variety of processes. This includes increased muscle mass, which occurs

through androgen-mediated myogenesis and muscle hypertrophy. Furthermore, muscle

AR plays an important role in regulating development of innervating motoneurons, as is

exemplified by the SNB neuromuscular system. Finally, recent evidence seems to link

AR in myocytes to SBMA incidence, suggesting a potential myogenic cause for disease

onset.

1.6. Androgens, AR and Fat

The anabolic effects of androgens in mediating muscle growth have mostly been

well elucidated and understood. Less well studied, however, is how androgens regulate

body fat. In males, androgens are largely thought to reduce overall body fat (Bhasin et al.,

1996). In females, the effects of T and other androgens on body fat are clouded by the

influence of estrogens. Teasing apart the effects of these two hormone classes is

paramount to understanding exactly how androgens are capable of modulating whole

body fat.

1.6.1. Sex Differences in Adiposity

Although much of the work in the field of mammalian sexual differentiation has

focused upon dimorphisms in reproductive organs and the nervous system, scientists are

now finding that there are significant differences between males and females in amount

Page 47: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

37

and distribution of body fat, as well as in the incidence of various metabolic disorders,

including obesity and T2DM. Women under the age of 50 have significantly reduced

likelihood of developing such diseases, but their prevalence increases markedly after the

onset of menopause (Ford, Giles & Dietz, 2002). Among adolescents, incidence of

metabolic disease is significantly higher among males than females (Ford et al., 2002).

Overall, the epidemiological data seems to suggest that ovarian hormones (ex: estrogens)

appear to be protective against metabolic disease because, prior to menopause, the

prevalence of these diseases is higher among males than females; however, after

menopause, females are more likely to develop these diseases (Shi, Seeley & Clegg,

2009). In addition, although older men are less likely to develop these diseases than older

females, older men are significantly more likely to do so than younger men. This suggests

a similar protective effect of androgens, since these hormones decline significantly with

age.

The increased risks due to obesity vary depending upon the location of adipose

accumulation (Bjorntorp, 1997). More specifically, concentration of adipose tissue

distributed in the abdominal (visceral) region carries a much greater risk for metabolic

disease than adipose accumulation in subcutaneous regions (Bjorntorp, 1997). This is

because different adipose depots have different properties. There are distinct sex

differences in adipose accumulation. Overall, females tend to have more subcutaneous fat

than males do (Lonnqvist, Thorn, Large & Arner, 1997), whereas accumulation of

adipose in visceral regions tends to occur to a greater degree in males (Mujica et al.,

2008). These effects also tend to change over the lifespan. As women age (particularly

after menopause, when estrogen levels decline), they tend to gain visceral adipose tissue.

Page 48: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

38

As men age, they also tend to gain visceral adipose tissue, although this gain has been

shown to be correlated with a reduced level of androgens in the bloodstream (Shi et al.,

2009). Thus, it appears that sex hormones do play a profound role in mediating adiposity

in both males and females, although this regulation is drastically different.

In males, androgens largely promote decreases in adiposity (i.e. reduction in

adipose tissue accumulation). Lines of evidence for this effect come from both treatment

of adult males with exogenous T, as well as natural reductions in T that occur during

aging. T supplementation decreases fat mass in both hypogonadal and eugonadal men

(Wilson, 1988; Bhasin et al., 2001). Furthermore, these effects of T on fat mass are

strongly correlated with the dose of T provided, as well as circulating levels in the

bloodstream (Bhasin et al., 1997; Snyder et al., 1999). These effects also appear to be

depot-specific, as T supplementation in older men preferentially reduces fat mass in

visceral depots (Marin et al., 1996). Studies of hypogonadal men seem to indicate similar

associations. A Finnish cohort study demonstrated that men who developed obesity in

late-adulthood had a 2.6-fold increased risk of having hypogonadism (Laaksonen et al.,

2005). Among men suffering from Klinefelter Syndrome (who have hypergonadotropic

hypogonadism), the prevalence of obesity and T2DM is raised by a factor of four, as

compared to age-matched controls (Bojesen et al., 2006; Ishikawa et al., 2008). Thus,

studies in humans seem to strongly suggest that androgens largely function to reduce

adiposity, primarily from visceral depots. Many therefore believe that the increase in

adiposity seen in aging men is directly related to age-related decline in androgen

signaling.

Page 49: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

39

The picture of sex hormones and adiposity is made significantly more complex

when one considers the role of estrogens (the female sex hormones). In order to more

fully contextualize the effects of androgens on adiposity, one must first understand how

estrogens contribute to this phenomenon. Similar to androgens, estrogens act upon their

own nuclear receptor (estrogen receptor – ER), which also functions as a transcription

factor. As mentioned previously, estrogens are derived from T, in a reaction mediated by

the enzyme aromatase. Significant evidence suggests that estrogens also function to

reduce adipose accumulation in females. In women, adipose accumulation increases

significantly after the onset of menopause, when circulation of estrogens declines

(Gambacciani et al., 1997), and these increases can be reversed by treatment of females

with exogenous estrogen (Haarbo, Marslew, Gotfredsen & Christiansen, 1991). This

evidence is supplemented by clinical investigation of women suffering from Polycystic

Ovarian Syndrome (PCOS). In PCOS, the ovary of the woman secretes abnormally high

levels of androgens, resulting in negative feedback on the hypothalamus and significant

suppression of estrogen secretion. 50% of women with PCOS are obese, demonstrating an

increase in visceral adipose tissue (Dunaif et al., 1987). While this was originally taken to

be evidence that androgens actually increase adipose tissue, it is now better understood

that the increase in adiposity is a consequence of reduced estrogen signaling in PCOS

patients. This postulation is supported by evidence from rodents. Ovariectomy of female

rats results in increased visceral fat, while administration of exogenous 17-β estradiol

subsequently reduces this adipose accumulation (Clegg, Brown, Woods & Benoit, 2006).

Furthermore, ablation of ERα results in increased adiposity and body weight in female

mice (Heine et al., 2000). Interestingly, knockout of ERα also affected adiposity in males,

Page 50: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

40

as male mice demonstrate increased fat accumulation in late adulthood. However, more

recent evidence suggests that the site-of-action for ER-mediated reduction in adiposity

appears to be the ventromedial hypothalamus, an area previously discussed to be a major

regulator of feeding behaviour. Selective knockdown of ERα in the hypothalamus

induces obesity and metabolic disease through hyperphagia (Musatov et al., 2007). Thus,

taken together, these studies demonstrate that action of estrogens have similar effects

upon adipose tissue as androgens. Furthermore, these effects do seem to be important in

both sexes, although to a larger degree in females. Lastly, it appears that estrogens

mediate their effects by regulating feeding behaviour, likely through interaction with

leptin (Asarian & Geary, 2002; Asarian & Geary, 2006).

1.6.2. Androgens and Adipocyte Development

While it appears that estrogens regulate adiposity by acting upon ERα in the brain

and modulating feeding, less is understood about how androgens exert their effects upon

adipose tissue. Generally speaking, reductions in adiposity are largely achieved through

two major methods (Shi et al., 2009). The first is through inhibition of adipocyte

differentiation and development. Adipocytes are derived from precursor cells (termed

‘preadipocytes’), and inhibition of their differentiation from these precursor cells would

subsequently reduce their accumulation. The second avenue by which to achieve a

reduction of adiposity is the more well-known mechanism of fat metabolism. Here

mature, developed adult adipocytes release triglyerides, which are lysed for the purposes

of deriving fatty acids. These released fatty acids subsequently undergo β-oxidation in

mitochondria of various tissues in order to harvest energy in the form of ATP.

Page 51: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

41

When it comes to androgens, it is largely believed that these hormones exert their

effects on adipose tissue through the first pathway. That is, androgens are largely believed

to inhibit the development of adult adipocytes from their precursor cells (Gupta et al.,

2008). Indirect evidence from animal models has suggested that high androgen levels

modulate the proliferation and differentiation of preadipocytes differentially in specific

fat depots (Garcia et al., 1999). The difficulty associated with studying precursor cells in

an in vivo setting provides a rationale for why this possibility has largely been probed

from an in vitro perspective. Nevertheless, several lines of evidence have supported the

idea that androgens are capable of inhibiting differentiation and development of

preadipocytes into mature adult cells. Treatment of epididymal (reproductive)

preadipocyte cells with either T or DHT inhibits the activity of an adipose-specific

isoform of the enzyme glyceraldehyde-3-phosphate-dehydrogenase (GAPDH)

(Dieudonne et al., 2000). Some work in this context on mature adipocytes shows that T-

treatment actually suppresses lipoprotein lipase (LPL) activity and lipid uptake, thus

inhibiting adipocyte growth. This provides a third potential mechanism by which

androgens might inhibit increased adiposity. Singh and colleagues (2003) found treatment

of mouse C3H 10T1/2 pluripotent cells (which are capable of differentiation into muscle,

fat, cartilage, and bone cells) with T reduced the number of mature adipocytes found. Pre-

treatment of cells with bicalutamide (a potent AR-antagonist) blocked the T-induced

inhibition of adipogenesis. Therefore, the preceding studies suggest that androgens are

capable of reducing adipose tissue by acting directly upon AR in preadipocytes and

inhibiting their development into mature cells.

Page 52: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

42

AR expression is well-documented in adipocytes and pre-adipocytes of both

humans (Dieudonne et al., 1998) and rodents (Dieudonne et al., 2000). It is believed that

AR’s function as a transcription factor may be harnessed in adipocytes and preadipocytes,

through downregulation of adipogenic genes (Matsumoto, Takeyama, Sato & Kato,

2003). Mature adipocytes treated with T demonstrate reduced expression of LPL and

GAPDH, which both promote adipocyte growth. Treatment of mouse C3H 10T1/2

pluripotent cells with T (Singh et al., 2003) reduced expression PPARγ2 mRNA and

protein, as well CCAAT/enhancer binding protein α (C/EBP). Both proteins are key

transcription factors necessary for adipogenic differentiation (Rosen & Spiegelman, 2000;

MacDougal & Mandrup, 2002). In a separate study, Singh et al. (2006) treated mouse

pre-adipocyte 3T3-L1 cells with T, and measured gene expression of various adipogenic

genes. Here, the scientists were able to recapitulate the downregulation of both PPARγ2,

as well as C/EBP. They also examined the Wnt gene family, which have been implicated

largely in the control of preadipocyte differentiation and adipogenesis (Cadigan & Nusse,

1997). It was found that activation of AR affected expression of various Wnt proteins in

the preadipocytes, suggesting a possible mechanism by which AR might modulate

adipogenesis. Once again, these effects were abolished by pretreatment of the cells with

bicalutamide. Overall, the evidence seems to suggest that androgen-mediated reduction in

adiposity occurs due to decreased expression of adipogenic factors by AR.

1.7. Where The Action Is: Lessons Learned from ARKO

Previous in vitro work concerning the role of AR in modulation of body

composition has demonstrated rather direct effects of AR action. As described previously,

Page 53: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

43

androgen-activation of AR has been shown to induce commitment of pluripotent cells to

the myogenic lineage (Singh et al., 2003). In addition, androgens have been shown to act

upon pre-cursor myoblast cells, promoting their development into mature myocytes

(Singh et al., 2009). Also, as mentioned, androgens have been shown to have opposite

effects on adipose, inhibiting differentiation of pluripotent stem cells into the adipogenic

lineage (Singh et al, 2006), and largely preventing development of pre-adipocytes into

mature cells (Gupta et al, 2008). Thus, these avenues of research have largely supported

the notion that androgens have direct effects upon their target tissues, and play a major

role in either promoting or inhibiting cellular differentiation and development.

While the previously mentioned studies have been fruitful in demonstrating in

vitro effects of androgens on various tissues (across various species), they are by

definition limited by the fact that they are conducted in isolation of other physiological

systems and cells. The physiology of mammalian organisms is inherently complex, with

tremendous interaction between various systems. For that reason, scientists ideally wish

to study physiological systems in vivo (i.e. in living organisms), in order to observe how

manipulations within one system can affect whole-body function. The advent of gene

targeting made such investigation possible. Traditional gene targeting uses homologous

recombination in order to alter a gene. Changes made include whole-gene deletion

(knockout), removal of portions of the coding region of a gene, addition of a gene

(transgenic technology), and induced point mutations. These genetic alterations can be

ubiquitous (i.e. occurring in all cells throughout the organism), or tissue-specific. With

regards to AR, gene targeting provides a powerful tool for understanding of sites of

androgen action. As discussed, AR expression is abundant in various tissues (Lee &

Page 54: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

44

Chang, 2003). By altering AR expression within a particular tissue (via either loss- or

gain-of-function), scientists can study the role of that tissue-specific AR, and its

contribution to various phenotypes (Kerkhofs, Denayer, Haelens, & Claessens, 2009).

1.7.1. Whole-Body Androgen Receptor Knockout

Using gene-targeting technology, the first androgen receptor knockout (ARKO)

mouse was produced in 2002 (Yeh et al., 2002). For decades prior to this advancement,

Testicular feminization mutation (Tfm) rats and mice had served as the classical animal

models in understanding loss of AR function, albeit primarily in males (Yarbrough et al.,

1990; Lyon and Hawkes, 1970). Since the AR gene is located on the X chromosome, and

is vital for male fertility, the scientists were unable to use traditional knockout

technology, and instead created a conditional knockout model. This was done through the

use of a cre-lox strategy. Such a system uses the expression of the P1 phage cre

recombinase (Cre) enzyme, which catalyzes the excision of DNA located between

flanking loxP sites (Holt and May, 1993). By using this methodology, researchers could

generate male founder mice carrying the floxed AR gene, who could then be mated with

female mice ubiquitously expressing Cre (through the use of the CMV promoter).

Resultant offspring would express the ARKO mutation in all tissues.

Upon examination of the initial ARKO mice, several striking phenotypes were

noted (Yeh et al., 2002). Similar to Tfm mice (Lyon and Hawkes, 1970), the external

genitalia of male ARKO mice show an ambiguous or feminized appearance. Agenesis of

the vas deferens, epididymis, seminal vesicles and prostate were reported, along with

significant decrease in the size of testes.

Page 55: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

45

Interesting findings were reported with regard to body composition in ARKO

mice. Yeh and colleagues (2002) found that cancellous bone volumes are lower in the

ARKO mice than in both female and male WT littermates. Furthermore, osteoclast (cells

involved in bone resorption) levels were higher in the ARKO mice, and they

demonstrated osteopenia. Similar effects of AR on bone resorption were reported by a

separate research group (Kawano et al., 2003). Here it was shown that whole-body

ablation of AR in males resulted in reduced trabecular and cortical bone mass, but no

change in bone shape. These findings were not found in ARKO females. Taken together,

these results provide strong evidence that androgen/AR action is important for

suppression of bone resorption.

Examination of skeletal muscle physiology in ARKO mice seemed to confirm

previously postulated ideas regarding the role of AR in muscle. As stated previously, it is

largely believed that androgens have anabolic effects on muscle mass, resulting in muscle

fiber hypertrophy (Ting and Chang, 2008). Others have suggested that, in addition to

mediating hypertrophy of individual muscle fibers, androgens act to promote myogenesis

and development/differentiation of mature myocytes from precursor cells (Singh et al.,

2009). MacLean and colleagues (2008) found marked change in skeletal muscle of

ARKO mice. Muscle mass is largely decreased in ARKO males, with no such differences

seen in ARKO females. Furthermore, muscle function appears to be impaired in ARKO

males. Isolated type II (EDL) muscles were found to demonstrate reduced maximum

tetanic force. This study largely demonstrates that peak skeletal muscle mass and function

in ARKO males is achieved through androgen/AR action.

Page 56: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

46

Perhaps most interesting of the phenotypes observed in ARKO mice relates to

adipose tissue and metabolism. The original study of ARKO mice found that overall body

weight in ARKO males closely resembled that of WT and ARKO females (Yeh et al.,

2002), with overall body fat and adipocyte size actually lower in the 8-week old mutant

males, as compared to WT brothers. These findings led the authors to postulate that AR

may actually be necessary for adipogenesis. However, these same scientists later recanted

this possibility once they had followed the male ARKO mice into late adulthood (Lin et

al., 2005). Aging ARKO males were found to demonstrate accelerated weight gain, even

overtaking WT males after 20 weeks of age. This weight gain resulted in excess adiposity

and obesity in later adulthood. Onset of obesity was also co-morbid with hyperglycemia,

progressively reduced insulin sensitivity and impaired glucose tolerance, suggesting that

adult ARKO males also develop T2DM. This late-onset obesity in ARKO males was

confirmed by a separate study (Fan et al., 2005). Here it was reported that ARKO males

also had lower levels of basal metabolism (as demonstrated through reduced oxygen

consumption), in addition to reduced rates of spontaneous activity. Taken together, the

above results all seem to suggest a model by which androgen activity is important for

reduction of adiposity, at least in late adulthood. T levels in males naturally decrease as

they age (Bhasin et al., 1996), which can partially account for increases in adiposity due

to aging (Shi et al., 2009).

1.7.2. Tissue-Specific Androgen Receptor Knockout

While the previously discussed research using ARKO mice has been fruitful, it

has largely just confirmed what has been believed about the role of androgens and body

Page 57: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

47

composition in males. That is, T is associated with increases in lean muscle mass, and

decreases in overall fat mass (Bhasin et al., 1996; Forbes et al., 1992). These studies still

do not indicate which tissues and cells are important for the effects of androgens on body

composition. An innovative way to tackle this problem is through the use of tissue-

specific AR knockout. One advantage behind the nature of ARKO mice (i.e. the floxed

AR gene) is that they can be mated with animals having tissue-specific Cre-recombinase

expression (Yeh et al., 2002). Such a mating would produce offspring that have null-AR

in the Cre-expressing tissue. This provides a very powerful tool for the study of

androgen-AR function in specific tissues. To date, these tissue-specific ARKO mice have

provided great insight into the distinctive roles of AR in mammary glands (Yeh et al.,

2003), reproductive tissue (Hu et al., 2004; Zhang et al., 2006; Wu et al., 2007) and the

immune system (Chuang et al., 2009; Lai et al., 2009).

While many questions remain regarding site-of-action for androgenic effects on

body composition, tissue-specific ARKO have provided significant insight in this realm.

Scientists first aimed to decipher the androgenic site for reduction of adipose tissue. As

discussed previously, it is largely believed that androgens are capable of reducing WAT

by acting upon AR in adipocytes themselves, inhibiting the differentiation of precursor

pre-adipocytes into mature cells, and down-regulating expression of adipogenic factors

(Singh et al, 2006; Gupta et al, 2008). Such a mechanism may account for the late-onset

obesity seen in male ARKO mice. To test this principle, ARKO mice were developed

with selective knockout of AR in adipocytes (aARKO; Yu et al., 2008). Interestingly,

body weight and adiposity of male aARKO mice did not differ from age-matched WT

brothers, and these animals did not develop obesity in later adulthood. This study

Page 58: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

48

revealed that the characteristic obesity seen in adult ARKO mice was not due only to loss

of AR in adipocytes. This issue was further probed by knockout of AR in hepatocytes

(hARKO; Lin et al., 2008). The liver is a major physiological site of fatty-acid oxidation,

and impaired hepatocyte function is associated not only with localized fatty-acid

accumulation (hepatic steatosis), but also obesity (Kammoun et al., 2009). However,

selective knockout of AR in hepatocytes also failed to recapitulate the obesity phenotype

found in whole-body ARKO mice. When the mice were put on a high-fat diet, hARKO

male mice developed hepatic steatosis and insulin resistance (likely as a consequence of

the steatosis), which was not found in WT brothers. These findings suggest that liver AR

may be important for local oxidization of fatty acids, but (similar to adipocyte AR) are

not sufficient in mediating androgenic reduction of adiposity.

To identify the role of AR in muscle, Ophoff and colleagues (2009), developed

ARKO mice lacking AR in myocytes (mARKO mice). Male mARKO mice demonstrate

reduced body mass and lean body mass (LBM). While there was a marked reduction in

size of the androgen-dependent levator ani (LA) muscle in male mARKO mice,

differences in other individual muscles were either very small or non-existent. Scientists

also found no differences in muscle contractile function, although they noted that there

was a significant conversion of muscle fiber type from fast to slow-twitch in male

mARKO mice. This study provided strong evidence that myocyte AR is important for

regulation and maintenance of skeletal muscle mass and fiber-type, but not function or

strength. Thus, taken together, tissue-specific ARKO has revealed that myocyte AR is

important for maintenance of mature skeletal muscle, but has been unable to identify a

site-of-action for androgenic reduction in adiposity.

Page 59: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

49

1.8. The Metabolic Potential of Muscle

Discussion of mammalian metabolism largely focuses upon organ systems as its

major players, including the brain, the liver, and the pancreas. For this reason, skeletal

muscle has been largely ignored as a prominent contributor to systemic metabolism.

However, in the last several decades, scientists have become acutely aware that changes

in skeletal muscle mass (and even atrophy/hypertrophy of individual fibers) can

drastically alter whole-body metabolism and energy homeostasis (Harrison & Leinwand,

2008). Muscle is a major storage site for glycogen, and a prominent target for insulin and

serum glucose uptake. Furthermore, as mentioned, catabolism of energy substrates by

skeletal muscle accounts for a significant proportion of systemic metabolism. We also

find that this metabolic capability is largely mediated by skeletal muscle mitochondria,

the oxidative energy builders within the cell.

1.8.1. Skeletal Muscle Metabolism

As briefly discussed previously, energy utilization in skeletal muscle is largely

dependent upon the muscle fiber-type in question. Type I (slow-twitch) muscle fibers are

characterized by a high mitochondrial content, while type II (fast-twitch) muscle have

less mitochondrial content, relying moreso upon glycolysis in order to meet energy

demands (Scott et al., 2001). In mammals, the type II fibers can be further subdivided into

the more oxidative type IIa fibers, and the more glycolytic type IIb fibers.

Muscle fibers require high levels of ATP in order to maintain their cycle of

contraction and relaxation (Hultman & Spriet, 1986). Skeletal muscle has three major

Page 60: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

50

methods by which it can form ATP. A majority of the ATP synthesized in muscle is

derived from muscle glycogen stores. First, glycogen can be directly broken down to

form ATP and lactic acid through the process of glycolysis. This method is relatively

inefficient, but can be performed quickly and in the absence of oxygen. It is the most

common method of energy utilization for type II fibers. Alternatively, ATP can be

derived through oxidative phosphorylation in muscle mitochondria. This process is

aerobic (requiring oxygen), but can derive far more ATP molecules per glucose molecule

than glycolysis. Additionally, it is capable of using fatty acids and amino acids as

substrates, while glycolysis is only capable of employing glucose. This type of energy

utilization is most common among type I fibers, although type IIa fibers use this method

to a larger degree than type IIb fibers. Finally, ATP can be synthesized in skeletal muscle

through phosphorylation of ADP by the molecule creatine phosphate (CP) and the

enzyme creatine kinase (CK) (Klivenyi et al., 1999). CP is created through

phosphorylation of the molecule creatine, which is derived from dietary amino acids, and

produced in the kidneys and liver. After production, it is subsequently transported into the

blood. Creatine is used almost exclusively by skeletal muscle, although a small

percentage of serum creatine is also transported to the brain and heart. After creatine is

taken up by skeletal muscle, it is phosphorylated to form energy stores in the form of CP.

When CP is broken, the released energy catalyzes the phosporylation of ADP to ATP.

While this process is relatively quick, it is inherently limited by the initial concentration

of CP in the cell. For this reason, weight lifters commonly use creatine supplements for

the purposes of increasing muscle energy, allowing them to lift more weight (Kadi et al.,

2008).

Page 61: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

51

The method of ATP synthesis used is largely dependent upon the intensity and

duration of muscle contraction. At moderate levels of skeletal muscle activity (and for

longer duration of exercise), most of the ATP used for muscle contraction is derived from

oxidative phosphorylation, and is driven by the breakdown of muscle glycogen stores into

glucose. As exercise progresses, and glycogen stores deplete, the muscles will turn to

fatty acid as substrate for energy utilization. As the intensity of muscle activity increases,

a greater fraction of total ATP production is formed by anaerobic glycolysis. Once muscle

activity is complete, CP and glycogen levels have largely decreased, and the muscle must

look to build up energy again by replenishing these stores.

The major source of energy supply in skeletal muscle comes from glycogen,

which are highly branched polysaccharide stores, composed of glucose subunits. The

uptake of individual glucose molecules by skeletal muscle is a highly specialized process,

which is mediated by the metabolic hormone insulin (Bouzakri et al., 2006). Synthesized

by the β cells of the pancreas, insulin is the major endocrine regulator of glucose

homeostasis, allowing for its uptake from the blood by peripheral organs, and facilitating

its storage as glycogen. Insulin functions by binding to the insulin receptor (IR), which

when activated results in phosphorylation of insulin receptor substrates (IRSs), eventually

facilitating transport of glucose across its transporter and into the cell. Expression of IR

and the major glucose transporter 4 (GLUT4) are readily found in skeletal muscle

(Holloszy, 2008). Once glucose enters the cell, it is subsequently packaged by the enzyme

glycogen synthase (GS) into glycogen. The activity of GS seems to be readily regulated

by insulin action, as IR knockout mice demonstrate reduced GS enzyme activity in both

skeletal muscle and brain (Araki et al., 1994). Once packaged into glycogen stores, they

Page 62: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

52

are ready to be utilized for ATP synthesis during periods of muscle contraction. Also, as

previously mentioned, fatty acids can serve as an important substrate for skeletal muscle

ATP derivation through oxidative phosphorylation. Fat is stored in the form of

triglycerides within specialized adipocyte cells. Triglycerides are broken down by the

enzyme LPL into glycerol and fatty acids, which are packaged with proteins into

aggregates called lipoproteins. This packaging allows for fatty acids to be transported

through the blood. Fatty acids enter muscle tissue through the fatty acid transport protein

(FATP; which also imports fatty acids into the mitochondria), and are then capable of

being used as a substrate for ATP production.

Muscle energy homeostasis may play an important role in the pathophysiology of

type 2 diabetes mellitus (T2DM), a major metabolic disease that is characterized by high

levels of serum glucose, and an inability for target tissues to uptake this glucose. In the

past, much research into T2DM has focused upon the β cell of the pancreas (where

insulin is produced), with β cell death being primarily linked to the disease. However,

scientists now understand that insulin resistance in target tissues may play as large of a

role in the development of T2DM as β cell failure (Biddinger & Kahn, 2006). In other

words, although the β cell is capable of producing significant levels of insulin, this

hormone may not be able to bind to the IR, or if it does, is incapable of generating the

signal cascade necessary for glucose transport. This leads to heightened levels of glucose

in the blood (characteristic of T2DM), as well as increased levels of insulin (a condition

known as hyperinsulinemia). Much of the research in this field has focused upon insulin

resistance in target tissues such as the brain and liver, but it is now clear that skeletal

muscle insulin resistance alone can result in T2DM. Insulin resistance is skeletal muscle

Page 63: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

53

largely occurs due to higher levels of fatty acids within the cell (Coen et al., 2010). Thus,

while fatty acids are an important substrate for the generation of ATP in skeletal muscle,

higher levels of fatty acids are capable of causing significant disruption of glucose

transport and subsequent glycogen synthesis (Turner et al., 2007).

Exactly how lipids accumulate within skeletal muscle myocytes is still a matter of

debate, and there are several potential mechanisms proposed. Largely, these mechanisms

can be grouped into two premises: (1) Increased fatty acid transport into the cell from the

blood or (2) Reduced fatty acid oxidation by muscle mitochondria for the generation of

ATP (or some combination of the two). It has been shown in humans (Bonen et al., 2004)

and rodents (Turcotte et al, 2001; Hegarty, Cooney, Kraegen, Furler, 2002) that

disruption of insulin signaling is induced by increased uptake of fatty acids into muscle.

This suggests that high levels of circulating triglycerides and lipoproteins may be

sufficient to induce insulin resistance, regardless of muscle oxidative capacity. A separate

body of evidence seems to suggest that defects in oxidative phosphorylation might

account for the increased fatty acid content of skeletal muscle myocytes (Hancock et al.,

2008).

1.8.2. Muscle Mitochondria

While nutrient transporters and membrane-bound receptors (and their subsequent

signaling targets) are known to play important roles in muscle metabolism, most research

is directed to the primary energy generator of the cell: the mitochondria. Even though

transport proteins and enzymes are important for packaging energy stores, the subsequent

breakdown of these stores and the derivation of ATP is largely accomplished by the

Page 64: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

54

mitochondria. This is especially true for type I (slow-twitch) muscle fibers, which rely

almost solely upon aerobic cellular respiration for energy for contraction.

Muscle mitochondria are a direct indicator of oxidative capacity in skeletal muscle

(Holloszy, 2008). As discussed previously, muscle fibers that rely upon oxidative

metabolism for ATP generation tend to be rich in mitochondria. Modulation of oxidative

capacity in adult skeletal muscle myocytes is largely achieved by two primary methods:

(1) mitochondrial biogenesis, and (2) mitochondrial enzyme activity (most notably the

enzymes of the electron transport chain and ATP synthase). Increasing either of these two

parameters will augment oxidative metabolism (create more ATP, while reducing the

amount of nutrient substrate available), while decreasing them will reduce oxidative

metabolism.

Increasing muscle mitochondrial content (mitochondrial biogenesis) occurs

naturally over the course of myocyte development. In mature adult cells, mitochondrial

content can be augmented through physical activity and exercise. There are several

consequences that arise from increased mitochondrial biogenesis (Holloszy, 2008). Not

only is the cell capable of generating greater amounts of ATP, there is also a significant

decrease in disturbance of energy homeostasis during times of profound muscle

contraction (such as seen during exercise). What this means is that there is a smaller

decrease in ATP and creatine phosphate consumed, as well as a smaller increase in ADP,

AMP and lactate produced (Constable et al., 1987). Furthermore, muscle fatigue is greatly

reduced (Dudley, Tullson & Terjung, 1987). Finally, and perhaps most interestingly,

proliferation of mitochondria is often linked with a change in substrate utilization toward

more oxidation of fatty acid, as opposed to breakdown of glucose, reducing fat stores

Page 65: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

55

throughout the body (Coggan et al., 1990; Hurley et al., 1986). Thus, increased

mitochondrial biogenesis not only alters methods of energy utilization within myocytes

themselves, but also improves whole-body systemic metabolism.

Various molecular players have been implicated in mediating muscle-specific

mitochondrial biogenesis and enzyme activity. The first major factor is linked to the

Peroxisome Proliferator-activated Receptor (PPAR) family. The PPAR nuclear receptors

(α, β, γ, δ) are activated by polyunsaturated fatty acids, and play an important role in fatty

acid homeostasis (Chawla et al., 2001). A specific co-activator of PPARγ, termed

peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), has become well

known as one of the significant modulators of mitochondrial biogenesis. Modifying PGC-

1α expression not only alters mitochondrial content, but also has significant effects on

whole-body metabolism and body composition. Muscle-specific overexpression of PGC-

1α results in large increases in functional mitochondria (Wu et al., 1998; Lehman et al.,

2000, Lin et al., 2002) and GLUT4 expression (Wende et al., 2007). PGC-1α mediates its

effects on mitochondrial biogenesis by docking on and activating the transcription factors

that regulate nuclear genes encoding mitochondrial proteins and that induce expression of

mitochondrial transcription factor A, which regulates mitochondrial DNA transcription

(Lin, Handschin & Spiegelman, 2005).

Another major cellular modulator of mitochondrial fatty-acid oxidation is the

enzyme 5’ AMP-activated protein kinase (AMPK), which is activated during muscle

contraction (Chen et al., 2003), as well as by external endocrine signals (Tamauchi et al.,

2002). The effects of AMPK activation are diverse in nature. In skeletal muscle, AMPK

functions by enhancing fatty acid uptake (Steinberg et al., 2004), and increasing fatty acid

Page 66: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

56

transport into mitochondria for β-oxidation (Munday, Carling & Hardie, 1988). It also has

pronounced effects upon glucose homeostasis in skeletal muscle, increasing GLUT4

content in the sarcolemma (Merrill, Kurth, Hardie & Winder, 1997). In these ways,

AMPK is capable of enhancing substrate utilization for ATP in skeletal muscle, reducing

serum glucose and fatty acids, and increasing systemic metabolism. AMPK has also been

shown to alter energy homeostasis through its effects on gene expression (McGee &

Hargreaves, 2010). Sufficient research shows that AMPK is capable of altering the

expression of several key genes in skeletal muscle metabolism. This includes genes

involved in angiogenesis, such as vascular endothelial growth factor (VEGF); and

glycolysis, such as hexokinase II (HKII), which phosphorylates glucose molecules in

preparation for energy harvest (Stoppani et al., 2002). AMPK also increases expression of

FATP, which increases transport of fatty acids into mitochondria (Barnes et al., 2005).

Substantial evidence also shows that AMPK is capable of increasing expression of

mitochondrial enzymes, particularly those involved in the aforementioned ETC

(Jorgensen et al., 2007; Garcia-Roves, Osler, Holmstrom & Zierath, 2008). Thus, taken

together, this work shows that AMPK plays an important role in mitochondrial biogenesis

and respiration in skeletal muscle.

Induction of regulators of skeletal muscle mitochondrial biogenesis tends to

increase metabolism in mammalian organisms. Overexpression of PGC-1α in skeletal

muscle fibers has several beneficial effects in aging mice (Wenz, Rossi, Rotundo,

Spiegelman & Moraes, 2009; Calvo et al., 2008). As compared to WT littermates at 20

months of age, these mice demonstrate reduced body fat, increased lean muscle mass, and

higher levels of basal metabolism. With regards to glucose metabolism, MCK-PGC-1α

Page 67: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

57

transgenic mice show increased insulin sensitivity and improved insulin signaling.

Similar findings have been demonstrated in the context of AMPK. In a key study, Ronald

Evans’ group showed that treatment of sedentary mice with the AMPK agonist AICAR

induced increases in oxidative capacity of skeletal muscle, fiber-type changes, and

induction of various metabolic genes (Narkar et al., 2008). They also found that these

mice show improved muscle-endurance when engaged in exercise, and show a synergistic

effect (in terms of muscle physiology and gene expression) as compared to mice that

exercised without the AMPK-agonist. These studies provide evidence of metabolic

benefits achieved through direct activation of mitochondrial biogenesis in skeletal

muscle.

While activation of these molecular players has been shown to have beneficial

effects on health, disruption of skeletal muscle mitochondria have been shown to result in

various pathologies. Mitochondrial myopathy (muscular disease characterized by

mitochondrial dysfunction) causes major disruption of metabolism and can result in

severe impairment or death (Johannsen & Ravussin, 2009). Several diseases (both

myogenic and neurogenic in etiology) that were originally thought to be due to unknown

mechanisms surrounding cellular toxicity are now thought to be related to mitochondrial

disease. For example, amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease,

characterized by motoneuron loss and muscular atrophy. Recent evidence has found that

mitochondria and impaired energy homeostasis may play a role in ALS (Menzies, Ince &

Shaw, 2002), with many ALS mouse models demonstrating skeletal muscle

hypermetabolism. Treating ALS mouse models with creatine (which, as described, is used

as an energy source in skeletal muscle) results in amelioration of ALS symptoms, and

Page 68: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

58

increased longevity (Klivenyi et al., 1999; Dupuis et al., 2004). Similar findings have

been shown in the context of Huntington’s Disease (HD), a neurodegenerative genetic

disease believed to be caused by neurotoxicity. Treatment of HD model mice with

creatine also has neuroprotective effects in aiding to slow progression and onset of the

disease (Ferrante et al., 2000). Interestingly, muscle biopsies taken from HD patients

show reduced expression of PGC-1α and lower levels of oxidative metabolism in isolated

myocytes (Chaturvedi et al., 2009).

PGC-1α-null mice demonstrate myopathy, coupled with reduced insulin-

sensitivity, T2DM and increased mortality (Lin et al., 2004), with similar deficits seen in

humans that lack the protein (Petersen, Dufour, Befroy, Garcia & Shulman, 2004).

Finally, myopathic symptoms can be attenuated by induction of transgenic PGC-1α

expression and subsequent mitochondrial biogenesis in skeletal muscle (Wenz, Diaz,

Spiegelman & Moraes, 2008). Skeletal muscle mitochondrial dysfunction has also been

implicated in pathogenesis of spinal and bulbar muscular atrophy (SBMA) (Ranganathan

et al, 2009; Musa, et al., In Prep). Thus, not only can augmentation of skeletal muscle

mitochondria improve metabolic parameters, but dysfunction of these organelles can also

result in manifestation of a wide variety of pathologies.

1.9. Objectives and Hypotheses

Identifying the site of action by which T mediates body composition has

significant implications for human health and medicine. The preceding discussion has

highlighted the fact that skeletal muscle is an important regulator of whole-body

metabolism, and changes within this tissue (whether gain or atrophy) have far-reaching

Page 69: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

59

effects on systemic metabolism. With that in mind, this study aimed to investigate

transgenic rats that overexpress AR in muscle fibers. Body composition was probed, with

special attention being paid to adipose tissue. Whole fat pads were dissected and analyzed

from animals, with specific differences in individual adipocytes noted. The necessity of T

for the effects seen was also investigated in transgenic and wild-type females. Finally, the

study aimed to discover whether differences in body composition could be pinpointed to

underlying differences in resting metabolism.

Since a loss-of-function knockout of AR in skeletal muscle fibers was sufficient to

induce muscular atrophy (Ophoff et al., 2009), it was hypothesized that overexpression of

AR in skeletal muscle fibers would increase skeletal muscle mass, likely by mediating

fiber hypertrophy. The importance of skeletal muscle to whole-body metabolism

suggested that an increase in muscle mass might be sufficient to increase resting

metabolism and reduce adiposity (Harrison & Leinwand, 2008). Thus, it was expected

that these transgenic rats would subsequently demonstrate reduced fat body mass, and

enhanced resting metabolism. Finally, it was unclear as to whether these effects could be

seen in transgenic females, since despite overexpression of AR in skeletal muscle, T

levels would be too low for any measurable differences to be noted. Therefore, it was

hypothesized that the differences found between transgenic and wild-type males could be

manifest in females through acute T-treatment.

Page 70: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

60

Chapter 2: Materials and Methods

2.1. Overview

The described study consisted of five separate experiments aimed at identifying

the role of muscle AR in regulation of body composition and metabolism. All 5

experiments used the rat species (Rattus norvegicus), while Experiments II and V

supplemented the work on rats by also employing the mouse species (Mus musculus). The

specific strains employed are discussed in further detail below. Since androgens (namely

T) have more prominent functions in males, most of the experiments (i.e. Experiments I,

II, IV and V) use this sex predominantly, if not exclusively. Female animals are also

investigated in Experiment II, while only Experiment III is performed exclusively with

females. A summary of all of the animals used is provided in Table 1.

Most of these studies involve investigation of skeletal muscle. For this purpose,

the extensor digitorum longus (EDL) muscle was typically examined. The EDL is often

used as a representative skeletal muscle since it is easily extracted, and its use has been

well characterized in studies of androgens and non-sexually dimorphic skeletal muscles

(Monks et al., 2006). Finally, subject genotyping as well as Experiment I involved the use

of various primer sequences, which are summarized in Table 2.

2.2. Animal Strains

2.2.1. HSA-AR Rats

HSA-AR transgenic rats were created and generously provided by Ligand

Pharmaceuticals Inc. (San Diego, CA). In these animals, overexpression of human AR is

Page 71: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

61

driven specifically in muscle fibers through the use of the Human Skeletal α-Actin (HSA)

promoter sequence. Expression of the α-skeletal actin (ACTA1) gene is largely restricted

to the striated muscles (more in skeletal muscle than cardiac) in all vertebrate species

studied, and is not found in major androgen target tissues such as the prostate or preputial

gland (Brennan & Hardeman, 1993). Similarly, androgens are capable of activating the

ACTA1 gene and promoter in isolated muscle cells, but not in liver, prostate or breast

cancer cells (Hong et al., 2008). Therefore, the HSA promoter is commonly used to drive

transgene expression specifically in skeletal muscle myocytes (Ex: Clapham et al., 2000;

Rao & Monks, 2009).

Generation of HSA-AR Tg rats was performed as described previously (Niel et

al., 2009). Human AR cDNA was obtained from a clone (as provided by Dr. S. T. Liao,

University of Chicago). A 3400-bp of the clone was ligated into a HSA expression

cassette, using BglII and ClaI restriction sites. The HSA promoter used was a fragment of

that found in the ACTA1 gene. Thus, the promoter used for development of this transgenic

strain was a truncated version of the full HSA promoter. The sequence and orientation of

obtained constructs was confirmed by sequencing. This was followed by pronuclear

microinjection of transgene DNA into Sprague-Dawley (SD) strain zygotes. Identification

of animals carrying the HSA-AR transgene was accomplished through PCR

amplification. Founders were subsequently backcrossed onto WT SD rats for at least 7

generations.

Initial characterization of HSA-AR rats was performed in order to determine

location and level of transgene expression (Niel et al., 2009). Reverse transcription

polymerase chain reaction (RT-PCR) using transgene-specific primers demonstrates

Page 72: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

62

expression of the transgene in limb muscle and BC/LA (sexually dimorphic skeletal

muscles) of neonates. No expression is found in muscle from WT neonates. Transgene

expression is also found in LA muscle of adult HSA-AR rats. RT-PCR was not performed

on non-sexually dimorphic muscle, nor was it performed on other tissue types. These

differences in mRNA expression of the transgene were also confirmed for protein

expression through western blot. AR was probed using a non-specific AR antibody

(probes for WT and Tg AR). In tissue taken from HSA-AR and WT adult rats, it was

demonstrated that the amount of AR protein was roughly 1.6 fold higher in LA muscle,

and 3.8 fold higher in EDL muscle of HSA-AR rats. Thus, taken together, it has been

shown that HSA-AR rats demonstrate higher levels of AR expression in skeletal muscle.

However, it remains unclear as to whether transgene expression is found in other tissue

types (including other muscle types) in HSA-AR animals. Functionality of the transgene

was assessed through gene expression of myoglobin, which is a typical marker of local T

action in skeletal muscle (Manttari, Anttila & Jarvilehto, 2008). Myoglobin mRNA levels

were found to be higher in EDL muscle of HSA-AR adult male rats, as compared to WT

male adults (Niel et al., 2009). This provides evidence of functionality of transgene-

derived AR.

2.2.2. Tfm Rats

For the last several decades, Testicular feminization mutation (Tfm) rodents have

provided the dominant model for understanding androgen insensitivity in mammalian

systems (Zuloaga et al., 2008). The Tfm phenotype is the product of a single-base pair

mutation in the AR gene (Yarbrough, et al., 1990). More specifically, it is the mutation of

Page 73: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

63

guanine to adenine within exon E, resulting in an amino acid change of arginine 734 to

glutamine within the ligand-binding domain of the AR gene. Thus, even though the AR

protein is successfully translated, due to the mutation in the ligand-binding domain, this

AR protein is not capable of binding to androgens, nor translocating to the nucleus to alter

expression of androgen-responsive genes. However, due to the simple nature of the

mutation (single amino acid alteration), it has been argued that there is some residual

sensitivity to androgens in Tfm rats, although it is believed to be largely negligible

(Zuloaga et al., 2008).

The translation of non-functional AR in Tfm rats results in a phenotype that is

analogous to that seen in human androgen-insensitivity syndrome (AIS) (Zuloaga et al.,

2008). Homozygous Tfm rats that are genetically male (i.e. possessing the XY

chromosome configuration) appear phenotypically female. They possess nipples typical

of female rats. Furthermore, their external genitalia demonstrate an ambiguous (intersex)

phenotype, with no discernible phallus. They are also infertile, due to defects in

spermatogenesis. Due to the X-linked nature of the mutation, only genetically male (XY)

carriers can demonstrate androgen insensitivity. Since Tfm males are infertile, obtaining

Tfm animals is therefore achieved by crossing a WT male with a heterozygous Tfm-carrier

(XTfmX) female.

Finally, it is important to discuss endocrine parameters in Tfm male rats. Most

notably, T levels appear to be extremely high in males, with serum concentrations

comparable to the high male range (Naess et al., 1976). This is presumably due to

uncoupling of HPG negative feedback. The mutation of AR in the hypothalamus means

that T will not have action on this region of the brain, and therefore will not be able to

Page 74: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

64

suppress secretion of LHRH or gonadotropins (FSH and LH). Concomitant with this

explanation, FSH and LH levels are also elevated in Tfm male rats (Naess et al., 1976).

This would account for the increased levels of T seen in these animals. Lastly, there have

been some differences noted in activity of the enzyme aromatase in Tfm rats. Recall that

aromatase is the enzyme that mediates the conversion of T into 17β-estradiol. Since

ArKO mice develop obesity (Fisher et al., 1998), in this context it is important to

understand any enzymatic differences between strains. In Tfm males, studies have found

impairment of aromatase activity in several parts of the brain, thus limiting conversion to

estrogens in these animals (Roselli, Salisbury & Resko, 1987; Rosenfeld, Daley, Ohno &

YoungLai, 1977). To date, differences in aromatase activity in other tissues are unclear.

Therefore, scientists must keep in mind the possibility that estrogen signaling may also be

impaired in males of this mutant strain.

2.2.3. HSA-AR/Tfm Rats

Obtainment of HSA-AR/Tfm animals is typically accomplished by breeding an

HSA-AR male with a heterozygous Tfm-carrier female. It could also be done through the

mating of a WT male with an HSA-AR/Tfm-carrier female. Resultant male offspring that

are homozygous for the Tfm mutation, but also express the HSA-AR transgene,

demonstrate a very interesting AR-expression phenotype. These animals possess non-

functional AR in all tissues (due to Tfm), except for muscle fibers, where AR is expressed

due to the transgene (Niel et al., 2009). As a result, these animals serve as a powerful

genetic model for investigation of AR function only in myocytes.

Page 75: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

65

Preliminary analysis of HSA-AR/Tfm males demonstrates increased expression of

AR in limb muscle fibers of neonatal rats, as compared to Tfm males (through

immunohistochemistry and confocal microscopy) (Niel et al., 2009). Phenotypically,

rescue of AR in skeletal muscle fibers of these rats does not promote development of the

sexually dimorphic SNB neuromuscular system in Tfm males. Post-natal maintenance of

perineal muscles (BC/LA) or innervating motoneurons is not found in this animal model,

suggesting that AR in skeletal muscle is not sufficient for development of this

neuromuscular system.

In this study, HSA-AR/Tfm male rats will be used to understand how AR within

muscle fibers alone can alter body composition and metabolism. For most purposes, they

will be compared and contrasted with Tfm littermates.

2.2.4. HSA-AR Mice

Transgenic mice overexpressing AR in skeletal muscle were developed by Monks

et al. (2007). Validation of the HSA promoter used for development of these mice was

performed through the creation of a LacZ (β-galactosidase) reporter strain, and it was

shown that the HSA promoter drives transcription only in skeletal muscle myocytes.

Transgenic HSA-AR mice were created using this promoter, driving expression of rat WT

AR cDNA. Obtained HSA-AR mice were then crossed with mice possessing the Tfm

mutation. Unlike in rats, Tfm mutation in mice is a single base deletion, resulting in a stop

codon (Zuloaga et al., 2008). For that reason, homozygous Tfm mice do not translate AR

protein. Subsequent HSA-AR/Tfm mice had tissues processed and were probed for AR

protein expression. Immunoblot assays confirm the specificity of the HSA promoter, with

Page 76: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

66

AR protein expression found in skeletal muscle only, and not in other tissues (such as

spinal cord, testes or heart).

Two major founding lines expressing the HSA-AR transgene were predominantly

used: L78 and L141 (Monks et al., 2007). There were significant differences in transgenic

AR expression found between these two lines. L141 animals have a significantly higher

transgene copy number (approximately 100 copies/pg RNA in L78, contrasted with

nearly 1000 copies in L141). Furthermore, L141 mice demonstrate significantly higher

AR expression in skeletal muscles, with differences noted in both AR mRNA and protein

level.

In terms of phenotype, marked differences also exist between the L78 and L141

lines. L141 mice demonstrate significant muscular atrophy, with male viability at birth

significantly reduced as compared to WT and L78 males. L141 male mice (or females

acutely treated with T) demonstrate reduced motor ability, body weight, and muscular

strength. Since the study described in this paper exclusively employs the L78 line, focus

will be paid towards the phenotype of this founding line. In contrast to L141 mice, L78

males are largely asymptomatic. Similarly, treating female L78 mice with exogenous T

does not result in muscular atrophy, as seen in L141 females. Body weight is reduced in

adult L78 males (as compared to WT brothers from the same line), but no defects in

motor functioning were noted. Some muscular pathology was noted in histological assays

of muscle fibers, with a reduced number of total EDL fibers recorded (as compared to

WT brothers from the same line). However, the degree of this pathology is significantly

reduced when compared to that seen in L141 mice.

Page 77: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

67

2.3. Subjects

2.3.1. Animals

All experiments described herein were accomplished using transgenic HSA-AR

and/or Tfm Sprague Dawley rats. HSA-AR and Tfm rats were also crossed in order to

produce the HSA-AR/Tfm strain, which expresses AR exclusively in myocytes.

Experiments II and V also employed transgenic L78 HSA-AR mice on a C57BL/6J

background. Please see Table I for breakdown of the number of animals used in each

experiment, and the genotypes of those animals. All animals were bred locally at the

University of Toronto at Mississauga. All procedures and experiments using live animals

were approved by the Office of Research Ethics at the University of Toronto, and adhered

to federal and National Institutes of Health guidelines. All animals were housed at

approximately 23oC, on 12h:12h light-dark cycles. Food and water were available ad

libitum.

2.3.2. Genotyping

Determination of animal genotype (including transgene expression or mutation)

was accomplished using polymerase chain reaction (PCR). At postnatal day (PND) 21,

animals were weaned from their mothers and housed in separate cages with food and

water. At this time, ear samples were taken by tissue puncture, placed in microcentrifuge

tubes on ice, and subsequently stored at -80oC for next-day lysis. The following day,

100µL of lysis buffer and 2µL of proteinase K were added to each microcentrifuge tube

containing a tissue sample. Samples were heated at 55oC for two hours, with brief

vortexing of samples after 30 minutes (to mix contents). Samples were then heated at

Page 78: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

68

95oC for 10 minutes, and spun in a centrifuge (10,000 RPM for 2 minutes) in order to

separate lysate from un-lysed tissue. This concentrated DNA was then diluted (1:10) in

distilled water for use in PCR.

PCR was performed using specific primers for each genotype. For HSA-AR rat

genotyping, DNA samples were analyzed using transgene-specific HSA-AR primers (F:

GGACAGGGCACTACCGAG; R: GGCTGAATCTTCCACCTAC), as well as primers

for the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH), used as

a control (F: ATGGGAAGCTGGTCATCAAC; R: GGATGCAGGGATGATGTTCT).

All PCR reactions include a known positive control, a known negative control, and a null

control. The reaction was run for a total of 31 cycles. Subsequent PCR product was run

on a 1.2% agarose gel for 30-35 minutes at 100 V. The gel was then removed and

visualized using ethidium bromide. HSA-AR is a 192 bp product, while GAPDH is a 440

bp product, creating two easily distinguishable bands. For genotyping of HSA-AR mice,

DNA samples were analyzed using transgene-specific HSA-AR primers (F:

AGTAGCCAACAGGGAAGGGT; R: GAGGCAGCCGCTCTCAGGGTG), with

primers for the housekeeping gene thyroid-stimulating hormone (TSH), used as a control:

(F: AACGGAGAGTGGGTCATCAC; R: CATTGGGTTAAGCACACAGG). The

reaction was run for 35 cycles. Subsequent PCR product was run on a 1.2% agarose gel

for 30-35 minutes at 100 V. The gel is then removed and visualized using ethidium

brominde. HSA-AR is a 650 bp product, while TSH is a 383 bp product, creating two

easily distinguishable bands.

Genotyping for the Tfm mutation was performed using PCR with primers for the

rat AR gene (F: GCAACTTGCATGTGGATGA; R: TGAAAACCAGGTCAGGTGC).

Page 79: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

69

Since this gene should exist in all rats, a housekeeping gene is not necessary. PCR

reaction ran for 35 cycles. Since the Tfm mutation is the result of a single base-pair

mutation, a portion of PCR product from each sample was digested using the restriction

enzyme Sau96I, which recognizes the 5’ GGNCC sequence. Tfm AR, due to the single

base pair mutation, cannot be cleaved by Sau96I, whereas WT AR should be cleaved into

two fragments. For each sample, restriction enzyme-digested PCR product was run next

to a non-digested PCR product (as a control). Gel electrophoresis was performed on a 3%

agarose gel for 30-35 minutes at 100 V. The gel was then removed and visualized using

ethidium bromide. All non-digested PCR product should demonstrate a single band,

signifying the AR gene. The presence of a single band in digested PCR product is

indicative of Tfm AR, while the presence of two bands indicates WT AR. Since

generation of homozygous Tfm females is not possible, the presence of only the non-

cleaved band in the digested-product is indicative of a Tfm male. Heterozygous Tfm-

carrier females can be identified from digested PCR product, through the presence of both

the single (non-cleaved) Tfm AR band, and the double WT AR band.

2.4. Experiment I: Tissue Specificity of Transgene Expression

Previous work on HSA-AR rats has demonstrated transgene expression (and AR

overexpression) in skeletal muscle (Niel et al., 2009). However, the fact that these

animals were produced using a truncated version of the HSA promoter implies the

possibility that transgene expression may be “leaky”, and found in other tissue types. For

this purpose, tissue specificity of transgene expression was analyzed.

Page 80: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

70

2.4.1. Reverse-Transcription (End-Point) PCR

At 16 weeks of age, WT and HSA-AR male rats were euthanized with an

overdose of sodium pentobarbital and dissected. EDL (skeletal muscle), heart (cardiac

muscle), urinary bladder (smooth muscle), kidney and white adipose tissue (WAT) were

removed. It is important to mention that separate tools were used for dissection of each

tissue, and were thoroughly cleaned after each animal, in order to limit any RNA

contamination. Tissues were removed, frozen in liquid nitrogen, and subsequently stored

at -80oC. Isolation of RNA was performed using TRIzol reagent. Tissues were slightly

degraded before being homogenized in the presence of 1 mL of TRIzol. Residual tissue

that could not be homogenized was discarded. Homogenized samples were allowed to

incubate for 5 minutes on ice before 200 µL of chloroform was added. Samples were

vortexed and components separated using centrifugation (14,000 RPM for 15 min). The

RNA exists exclusively in the colourless upper aqueous phase, from where it was

isolated. The remaining organic phase was discarded. Precipitation of RNA pellets was

accomplished by addition of isopropyl alcohol, manual shaking, and incubation at room

temperature for 2 hours. Following incubation, component separation through

centrifugation (12,600 RPM for 10 min) was carried out, resulting in the formation of an

RNA pellet. Next was an RNA wash phase, where the supernatant was removed, and the

pellet was washed with 1 mL of 75% ethanol. Centrifugation was carried out once again

(7,600 RPM for 5 min), after which point ethanol was removed. Finally, RNA was re-

dissolved. The pellet was allowed to briefly air-dry, before being washed in

diethylpyrocarbonate (DEPC) water (approximately 25 µL, potentially more if the pellet

could not be dissolved). Following this, the isolated RNA was stored at -80oC.

Page 81: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

71

After successfully isolation, RNA samples were analyzed for stability (non-

degradation) through glyoxylation and electrophoresis. Stable RNA was next treated with

DNase in order to remove any residual DNA. Next, the RNA was converted to cDNA

through a reverse transcriptase-mediated reaction. Newly acquired cDNA was then used

in a RT-PCR reaction (35 cycles) with transgene-specific primers for human AR (F:

AGGAAGCAGTATCCGAAGGCA; R: GGACACCGACACTGCCTTACA). Expression

of the housekeeping gene GAPDH was analyzed in a separate reaction (using previously

described primers), in order to act as a control. PCR products were electrophoresed on a

1.5% agarose gel. Presence of an electrophoresed band would be indicative of gene

expression. Analysis was performed for both WT (n = 3) and HSA-AR (n = 3) male rats.

2.5. Experiment II: Body Composition Analysis

Since androgens have been shown to have significant effects on body composition

(namely increasing lean muscle mass and decreasing fat mass), this possibility was

investigated in rat strains (WT, HSA-AR, Tfm, HSA-AR/Tfm), as well as comparative

mouse strains (WT, L78 HSA-AR).

2.5.1. Dual-Energy X-Ray Absorptiometry

Body composition was analyzed using dual-energy X-ray absorptiometry (DXA)

scanning. DXA scanning is a common method of measuring body composition, and is

employed in basic science animal studies, as well as in clinical contexts with patients

(Malina, 1969). These scans emit X-rays, which are directed at the subject, and

subsequently absorbed by the tissues of the body. Different tissue-types (namely lean

Page 82: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

72

mass, fat mass, and bone) absorb X-ray emissions to different degrees. Using these

differences in absorption, the machine is able to derive estimates of total body mass, as

well as the mass of each individual component of body composition.

Animals were scanned using a standard DXA machine (Hologic QDR4500;

Hologic Inc., Waltham, MA). For each DXA session, the machine was first calibrated

through the use of standardized phantom models. Measured parameters of these phantom

scans were maintained across sessions, in order to ensure the comparative validity of

scans performed on different days. Before scanning, animals were anaesthetized using

isoflurane, and maintained under anesthesia for the duration of the experiment. DXA

small animal scans take approximately 2 minutes each.

For rats, analysis was performed on WT males (n = 11), WT females (n = 8),

HSA-AR males (n = 11), HSA-AR females (n = 9), Tfm males (n = 7), Tfm-carrier

females (n = 7), HSA-AR/Tfm males (n = 9) and HSA-AR/Tfm-carrier females (n =7).

Rats were group-housed, with 3-5 animals per cage. Males and females were caged

separately. Rats were scanned bi-weekly, beginning at 4 weeks of age, and ending at 10

weeks of age. For mice, analysis was performed on the L78 line males: WT (n = 5) and

HSA-AR (n = 6). Mice were measured once by DXA at 24 weeks of age.

2.5.2. Parameters Examined and Derived

DXA analysis provides the operator with estimates of various parameters of body

composition. The includes total body mass, bone mineral content (BMC; the total amount

of bone mass), bone mineral density (BMD; the amount of bone per unit of bone area),

total fat body mass (FBM; the amount of adipose mass on the body), lean mass + bone

Page 83: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

73

mineral content (the total amount of non-FBM on the body), and FBM% (the percentage

of total mass accounted for by fat). Given these parameters, lean body mass (LBM) could

be derived. LBM was derived by taking the Lean+BMC parameter and subtracting the

known BMC value. LBM% was then determined using this total LBM and the known

body mass. For all scans, animal body weight was measured using a weighing scale, and

compared with the provided total body mass from the DXA scan, in order to validate

DXA accuracy. It was found that the measured weight by DXA never differed from the

actual mass by more than 2%, suggesting that the DXA machine was highly accurate in

its measures.

2.5.3. Dissections and Tissue Weights

In order to determine if the findings from DXA analysis could be confirmed in

individual tissues, dissections were carried out to measure weights of individual muscles

and fat pads of WT, HSA-AR, Tfm and HSA-AR/Tfm male rats. At 16 weeks of age, rats

were euthanized through overdose with sodium pentobarbital. Individual EDL and AT

muscles were dissected and weighed. WAT weight was measured by removal of

perigonadal (reproductive) fat pads from rats. Dissections were carried out for WT (n =

10), HSA-AR (n = 12), Tfm (n = 7) and HSA-AR/Tfm (n = 10) males.

2.6. Experiment III: Testosterone Treatment of Adult Females

This experiment attempted to recapitulate effects of the transgene seen in HSA-

AR male rats through acute treatment of adult HSA-AR female rats with exogenous T.

This was a within-group study, with all animals having body composition measured

Page 84: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

74

during treatment with T, as well as when T capsules were removed (and replaced with

vehicle capsules). Analysis was carried out on WT (n = 8) and HSA-AR (n = 9) female

rats, aged 24-30 weeks.

2.6.1. T-Capsule Surgeries

Treatment of females with exogenous T was done through implantation of silastic

capsules containing the hormone. This methodology has been used before in many

studies, and is a standard method of treating animals with exogenous hormone (Monks,

Vanston & Watson, 1999; Monks & Watson, 2001; Monks et al., 2001a; Monks et al.,

2001b). T-capsules containing exogenous T are SILASTIC brand (1.57mm inner diameter

X 3.18mm outer diameter, Dow Corning, Midland, MI), containing T (20 mm in length,

Steraloids, Newport, RI). After baseline DXA scanning, all animals were anaesthetized

with isoflurane. A small incision was made on the dorsal side of the rat, and two T

capsules were inserted subcutaneously (just beneath the skin). Incisions were

subsequently stitched and sealed, and animals were treated with an analgesic (Anafen) to

relieve any pain. Animals were also single-housed for three days after surgery, in order to

ensure proper closure of the wound. Following 4 weeks of exogenous T-treatment, all

animals were returned to surgery and anaesthetized again with isoflurane. The dorsal

incision was re-opened, and the T capsules were removed. Capsules were checked to

ensure that excess T had not been released. After T capsules had been removed, they were

replaced by two vehicle capsules (Silastic capsules that were empty), and the incision was

then sealed and stitched as described above.

Page 85: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

75

2.6.2. Body Composition Analysis

Female rats were subjected to DXA scanning before T treatment, in order to

establish baseline measures of body composition. Following T treatment, animals were

scanned weekly for 4 consecutive weeks. Immediately following the final scan of this

regimen, T capsules were removed, and vehicle capsules replaced, as described above.

Females were then scanned by DXA weekly for 4 consecutive weeks, in order to

determine if and how subsequent T withdrawal would alter body composition in HSA-AR

females.

2.7. Experiment IV: Adipose Histology

In order to determine effects of muscle AR on WAT at a cellular level, histology

of adipose tissue was conducted to examine the size of individual adipocytes.

2.7.1. Sample Preparation and Sampling Strategy

At 16 weeks of age, animals were euthanized through overdose with sodium

pentobarbital and dissected. Reproductive perigonadal fat pads were dissected in the same

way from all animals. Following dissection, removed fat pads were placed in 10%

phosphate-buffered formalin (PBF) for fixation for at least 5 days. Following fixation, a

sample of the whole fat pad was used to perform histology. This sample was taken from

the same part of the fat pad in all animals. Perigonadal fat pads are directly attached to the

testes in males, and so the sample was removed from the part of fat pad furthest from the

testes. After fixation, adipose samples were processed to prepare for paraffin embedding.

Samples were stored in individual plastic cassettes and placed in an automated basket,

Page 86: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

76

which takes the cassettes through each solution. Tissue was incubated in various solutions

(distilled water, 70% ethanol, 70% ethanol, 95% ethanol, 95% ethanol, 100% ethanol,

100% ethanol, xylene, xylene, paraffin, paraffin) for 1 hour per solution. Following

processing, tissue samples were properly oriented and embedded into paraffin blocks

using molds. The tissue was allowed to set overnight before sectioning.

Paraffin blocks were sliced on a microtome at room temperature, and at a

thickness of 6µm. Sections were mounted on slides using gelatin solution. Slides were left

to dry overnight on a slide heater. The following day, the slides were stained. Slides were

first cleared in xylene for 13 minutes (to remove paraffin), followed by dehydration in

graded alcohols (100%, 100%, 95%, 70%) for 3 minutes each. Slides were washed with

distilled water for 3 minutes, before being incubated in hematoxylin (Sigma-Aldrich,

Oakville, Ontario, Canada) for 5 minutes. Following hematoxylin staining, slides were

washed in distilled water for 1 minute, before being processed with eosin and

subsequently coverslipped.

Resultant sections needed to be analyzed systematically in order to maintain

consistency across subjects. Images were acquired using an Olympus bright-field

microscope (model BX51; Olympus, Tokyo, Japan), a 4X objective, and a colour

videocamera (Cool Snap Pro Color; Roper Scientific, Duluth Georgia) with Image Pro

Plus Software (Media Cybernetics Inc., Silver Spring, MD). Images were taken at 40X

magnification, at a resolution of 680 X 512 pixels. For each adipose cross-section, three

photomicrographs were taken for analysis. All photomicrographs were taken in the same

area of the cross-section for all subjects, in order to maintain consistency.

Page 87: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

77

2.7.2. Measurements

In order to quantify adipocyte size, photomicrographs were imported into ImageJ

software (National Institutes of Health, Bethesda, MD), which was used to trace cell size.

Calibration of the software was performed prior to measurements of size, so as to provide

accurate area measures in µm2. Due to the nature of the hematoxylin and eosin stain, not

all individual adipocytes were clearly demarked. For that reason, only cells whose

complete membrane border could be seen were subsequently traced and analyzed. For

each animal, a minimum of 150 measures were taken, which were then averaged in order

to determine the mean adipocyte size. Analysis was conducted on WT (n = 6), HSA-AR

(n = 6), Tfm (n = 6), and HSA-AR/Tfm (n = 4) males.

2.8. Experiment V: Energy Balance and Metabolic Analyses

We investigated parameters of energy expenditure in order to determine if any

changes in whole body composition might be associated with alterations in these

parameters.

2.8.1. Resting Metabolism by Indirect Calorimetry

Evaluation of resting metabolism is typically accomplished through the use of

indirect calorimetry, which uses indicators of metabolism in order to derive an estimate of

RMR. The most common method of indirect calorimetry is oxygen (O2) consumption.

Consumption of oxygen is closely related to metabolism, as O2 molecules are required by

cells undergoing oxidative phosphorylation. O2 is reduced by the transfer of two electrons

from cytochrome C oxidase in order to produce two water (H2O) molecules and is a

Page 88: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

78

necessary step in the production of ATP. For that reason, higher levels of O2 consumption

are indicative of higher rates of metabolism (Holloszy, 2008).

At 12 weeks of age, metabolic measures were taken from male rats through

analysis of O2 consumption. Animals were placed inside a 700 mL (8.5 cm diameter, 13.0

cm length) cylindrical gas-exchange chamber (model G114; Qubit Systems, Kingston,

Ontario, Canada). Calibration of the system was first achieved by passing nitrogen gas

through the system (which provides a “0” O2 concentration baseline level). Room air was

then pumped through the chamber at a flow rate of 400 mL/min, and outflow O2

concentration was measured by a flow-through oxygen analyzer (model S102; Qubit

Systems). Concentration of outflow O2 was analyzed and displayed by gas-exchange

software (Logger Pro version 3; Vernier Software, Beaverton, OR). Animals were kept in

the chamber until outflow O2 concentration levels were stable, which was defined by no

deviation in concentration of more than 0.02% over a period of 30 minutes. The

difference between final outflow O2 concentration and inflow concentration (i.e.

concentration of O2 in room air) was used to determine oxygen uptake (in µL/min).

Animals were matched for time of day at which the measures were recorded. In order to

derive RMR in (J/g min), values of O2 uptake were corrected for body mass. Indirect

calorimetry was measured for WT (n = 10), HSA-AR (n = 12), Tfm (n = 7), and HSA-

AR/Tfm (n = 10) male rats. Indirect calorimetry was also measured for WT (n = 5) and

HSA-AR (n = 6) L78 male mice, aged 22-23 weeks.

Page 89: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

79

2.8.2. Spontaneous Activity Measures

Animals were measured bi-weekly, starting at the age of 4 weeks until the age of

10 weeks. These ages were chosen as they correspond to the periods at which body

composition was measured in male rats. Locomotor activity was assessed in clear, acrylic

glass boxes (L 43 X W 22 X H 25 cm), with clear, ventilated acrylic glass lids. The boxes

were separated by opaque screens, which prevented the rats from seeing one another

during testing, and thus eliminated visual conspecific cues. Each activity box was

equipped with two arrays of 16 X 16 infrared photo-beams, spaced 2.5 cm apart

(constructed by the Centre for Addiction and Mental Health, University of Toronto,

Toronto, Ontario). The bottom array, positioned 3 cm above the floor of the chamber,

recorded horizontal movement. The top array, positioned 15 cm above the floor of the

chamber, recorded rearing behaviour. These arrays were connected to a computer via an

interface that detected interruptions in the photo-beam (i.e. beam breaks induced by

activity). Each activity session lasted for 60 minutes, and animals were counterbalanced

for the time of day at which they were measured. Activity measures were calculated for

WT (n = 4), HSA-AR (n = 6), Tfm (n = 3) and HSA-AR/Tfm (n = 5) male rats.

2.9. Statistical Analyses

Experiments II, IV and V (between-group studies involving HSA-AR and Tfm

rats) were analyzed using two-way multivariate ANOVA, with HSA-AR and Tfm as

between-subject factors (for body composition analysis, an ANOVA was calculated for

each week that animals were tested, and all between-group comparisons were made at

each age). Differences between groups were analyzed using independent sample student

Page 90: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

80

t-tests. Parts of experiments II and V that employed L78 mice were analyzed using

independent sample student t-tests to compare WT and HSA-AR groups.

Experiment III, which involved T-treatment of WT and HSA-AR adult female

rats, was a within-group study, in which both groups were exposed to both treatments (T

and vehicle). Data was examined using a repeated-measures ANOVA with HSA-AR as a

between-subject factor and total body mass, LBM%, FBM%, raw LBM or raw FBM as

the within-subject factor. Statistical analyses were run separately for the first 5 weeks

(baseline measures and 4 weeks of T-treatment) and for weeks 4-8 (final week of T-

treatment, and 4 weeks following removal of T). Pairwise comparisons were performed

using unprotected paired t-tests if an interaction was found between the within-subject

and between-subject factors. Otherwise, a Dunnett correction for family-wise error was

applied. Pairwise comparisons were made with each week either being compared to

baseline (applies for weeks 1-4) or week 4 (applies for weeks 5-8). The use of these two

epochs would allow for evaluation of effects of T-treatment (for the former weeks), and

subsequent T withdrawal (for the latter weeks). For all studies, α was set at P < or = 0.05.

Complete statistical analyses are included in Appendix A.

Page 91: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

81

Chapter 3: Results

3.1. Overview

HSA-AR animals were characterized to determine tissue specificity of transgene

expression. Subsequently, male and female rats (HSA-AR and WT) were analyzed for

changes in body composition. HSA-AR animals were generally compared against WT

littermates on the same Tfm background (i.e. comparisons were largely WT vs. HSA-AR,

and Tfm vs. HSA-AR/Tfm). This is due to the fact that Tfm male rats demonstrate

significantly reduced body mass, which strongly confounds any analyses of body

composition made when comparing these rats to WT males. After discovering differences

in body composition of male HSA-AR rats, but not females, these effects were induced

by acute T-treatment of HSA-AR females. Finally, differences in overall energy balance

were investigated, in order to gain some understanding of the physiological mechanism

behind the differences seen.

Where available, studies in rats were supplemented with similar experiments in

HSA-AR mice. This would allow for investigation of the generality of the effects seen

(i.e. whether they are found in other species).

3.2. Experiment I: Tissue Specificity of Transgene Expression

Due to the fact that HSA-AR rats were generated using a truncated version of the

full HSA promoter, it was necessary to qualitatively analyze various tissues of these rats

in order to determine if they expressed the transgene.

Page 92: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

82

3.2.1. Transgene mRNA is Expressed in Muscle Tissue of HSA-AR Animals

Dissected tissues from adult (16-week old) WT and HSA-AR male rats were

analyzed using RT-PCR and transgene-specific primers (Fig 1). EDL (skeletal muscle),

urinary bladder (smooth muscle). heart (cardiac muscle), WAT and kidney were all

examined for both genotypes. RT-PCR found no expression of the transgene in any

tissues taken from WT animals. Conversely, transgene mRNA expression was found in

skeletal muscle, smooth muscle, and cardiac muscle of HSA-AR males. No transgene

expression was demonstrated in adipose tissue or kidney of HSA-AR males. Thus, HSA-

AR mRNA expression seems to be found in myocytes of HSA-AR animals only.

3.3. Experiment II: Body Composition Analysis

Bi-weekly DXA scanning was performed on WT, HSA-AR, Tfm and HSA-

AR/Tfm rats (from 4 weeks of age until 10 weeks), in order to determine whether AR in

muscle is capable of modulating body composition. Since T secretion increases markedly

at the onset of puberty (roughly 6 weeks in rodents, see Lee & Chang, 2003), examining

these time points would allow for some delineation of T-dependency. If the action of

post-natal T on the transgene did indeed affect body composition, such differences likely

should not arise prior to the onset of puberty. Various parameters were examined,

including whole body mass, lean body mass (LBM), LBM%, fat body mass (FBM),

FBM%, bone mineral content (BMC) and bone mineral density (BMD). Representative

images of male rats (scanned at 10 weeks of age), including WT (Fig 2a), HSA-AR (Fig

2b), Tfm (Fig 2c) and HSA-AR/Tfm (Fig 2d) are shown. Due to the large amount of

statistical analyses carried out in examination of body composition, only pertinent

Page 93: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

83

statistics will be presented in text. This largely relates to main effects as determined by

two-factor (HSA-AR and Tfm) ANOVA. For complete statistical analyses (including

results of all ANOVA and between group t-tests), please refer to Appendix A.

3.3.1. HSA-AR Expression Does Not Regulate Body Mass in Rats

We first used DXA scanning to determine if the HSA-AR transgene could alter

whole body mass in male rats (Fig 2e). Interestingly, no differences in total body mass

were found due to HSA-AR. This was the case for males at 4 weeks (F3,25 = 0.359, P =

0.553), 6 weeks (F3,40 = 0.408, P = 0.528), 8 weeks (F3,34 = 1.181, P = 0.286) and 10

weeks (F3,34 = 3.339, P = 0.077). As expected, a main effect of Tfm was found to

influence whole body mass. No effect was found at 4 weeks (F3,25 = 0.817, P = 0.373),

however Tfm males were found to have lower body mass than non-Tfm males at 6 weeks

(F3,40 = 4.517, P < 0.05), 8 weeks (F3,34 = 20.641, P < 0.001) and 10 weeks (F3,34 =

22.116, P < 0.001). This was not surprising, as Tfm males develop an intersex phenotype,

and are found to have lower overall body weight (Zuloaga et al., 2008). No interaction

was found between HSA-AR and Tfm for any age.

3.3.2. Increased Lean Muscle Mass Percent in HSA-AR Male Rats

Selective knockout of AR in myocytes alone has been shown to be sufficient to

induce muscular atrophy in mice (Ophoff et al., 2009). With this in mind, we investigated

lean muscle mass of our HSA-AR rats using DXA scanning. We hypothesized that

overexpression of AR in myocytes should increase lean muscle mass. We first compared

LBM%, which is the total amount of lean mass per unit of total body mass (Fig 2f). It was

Page 94: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

84

found that HSA-AR is capable of increasing LBM%. At 4 weeks, no effect of HSA-AR is

found (F3,25 = 1.219, P = 0.278). However, effects become significant after the onset of

puberty at 6 weeks (F3,40 = 17.748, P < 0.001), as well as 8 weeks (F3,34 = 10.630, P <

0.01) and 10 weeks (F3,34 = 6.510, P < 0.05). Unexpectedly, we found a similar effect of

Tfm in increasing LBM%. Similar to the main effect of HSA-AR on this parameter, no

effect of Tfm is seen at 4 weeks of age (F3,25 = 0.205, P = 0.654), but mutation increases

LBM% at 6 weeks (F3,40 = 4.396, P < 0.05), 8 weeks (F3,34 = 27.393, P < 0.001) and 10

weeks (F3,34 = 11.549, P < 0.01). This finding was surprising as it was contrary to the

previously cited literature on mARKO mice (Ophoff et al., 2009), as well as the muscular

atrophy noted in whole-body ARKO (MacLean et al., 2008). Here it seems that lack of

functional AR may potentially result in increasing LBM%. No interaction between HSA-

AR and Tfm was found for any age.

Interestingly, the effects due to HSA-AR and Tfm do not directly carry over when

examining raw muscle mass. Here, raw LBM values are provided by DXA, but not

corrected for overall body weight (Fig 2h). No significant effect was found for HSA-AR

on LBM at any age measured. Pair-wise comparisons between groups on the same genetic

background (i.e. WT vs. HSA-AR and Tfm vs. HSA-AR/Tfm) also demonstrate no

difference. As expected, there is a main effect of Tfm, with Tfm males having lower levels

of raw LBM at 8 weeks (F3,34 = 44.909, P < 0.001) and 10 weeks (F3,34 = 11.560, P <

0.01). This was not surprising as Tfm males have lower overall body mass, when

compared to WT littermates, and thus would be expected to have lower levels of raw

LBM. Once again, no interaction was found between HSA-AR and Tfm.

Page 95: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

85

In summary, these results suggest a disparity between HSA-AR and WT animals

in LBM%, with HSA-AR animals having higher levels. However, this difference in

proportion of LBM is not accounted for by simply increased LBM, suggesting that other

components of body composition may additionally be altered. We also demonstrate an

interesting effect of Tfm in increasing LBM%, which was contrary to expectations.

3.3.3. Reduced Fat Body Mass in HSA-AR Male Rats

With the existing disparity between WT and HSA-AR rats demonstrated in

LBM%, we investigated body fat to determine if this component of body composition

might be altered due to transgene expression. Whole-body ARKO male mice develop

increased adipose tissue and eventual obesity (Lin et al., 2005; Fan et al., 2005),

suggesting that androgens and AR can mediate this major energy storage site. This effect

cannot be recapitulated by knockout of AR in adipocytes (Yu et al., 2008) and

hepatocytes (Lin et al., 2008). Unexpectedly, knockout of AR in myocytes was found to

decrease fat mass (Ophoff et al., 2009), but the authors were unable to provide a

satisfactory explanation for this effect. Thus, whether myocyte AR is capable of

modulating fat body mass (FBM) is currently unknown.

Investigation of FBM% (total amount of fat mass per unit of whole body mass) in

male rats shows a main effect of the transgene in reducing this parameter (Fig 2g).

Similar to effects on LBM%, no differences are seen at 4 weeks of age (F3,25 = 1.200, P =

0.282). However, decreased FBM% is noted in HSA-AR rats at 6 weeks (F3,40 = 16.275, P

< 0.001), 8 weeks (F3,34 = 10.004, P < 0.01) and 10 weeks (F3,34 = 6.360, P < 0.05). Once

again, Tfm is found to act similarly to HSA-AR in improving body composition by

Page 96: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

86

reducing FBM% in male rats. Effects emerge at 6 weeks (F3,40 = 4.718, P < 0.05), and are

also seen at 8 weeks (F3,34 = 11.640, P < 0.01) and 10 weeks (F3,34 = 12.837, P < 0.001).

Unlike the differences seen in LBM%, the effects on FBM% carry over when one

examines raw FBM (not corrected for weight; Fig 2i). That is, HSA-AR male rats

actually demonstrate reduced levels of FBM as compared WT littermates. Similar to other

effects, no differences are seen at 4 weeks (F3,25 = 2.216, P = 0.147), but are present at 6

weeks (F3,40 = 10.541, P < 0.01), 8 weeks (F3,34 = 7.769, P < 0.01) and 10 weeks (F3,34 =

6.662, P < 0.05). Tfm also reduced raw FBM, as adiposity is lower in male rats expressing

this mutation at 6 weeks (F3,40 = 4.718, P < 0.05), 8 weeks (F3,34 = 11.640, P < 0.01) and

10 weeks (F3,34 = 12.837, P = 0.001). This is to be expected, due to lower levels of whole

body mass found in Tfm males, as compared to WT littermates.

These results tell us that transgene expression reduces FBM in male rats (both raw

fat, and when corrected for body weight). Similar main effects are seen due to Tfm, with

mutant males demonstrating reduced FBM% (as well as lower raw FBM value, which

was expected due to lower overall body mass in the mutants). This finding was surprising,

as it identifies a novel role for muscle AR in reducing FBM.

3.3.4. No Effects on Body Composition of HSA-AR Female Rats

We also examined body composition of WT, HSA-AR, Tfm and HSA-AR/Tfm

female rats. It was not expected that the transgene would have pronounced effects on

females, due to significantly reduced levels of circulating T in this sex.

Analysis of whole body mass showed no differences between groups at any time

point (Fig 3a). Females were also investigated for effects on LBM% (Fig 3b), and FBM%

Page 97: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

87

(Fig 3c). No significant differences were found in body composition between females at 4

weeks, 8 weeks or 10 weeks (see Appendix A for complete statistics). Interestingly, a

transient effect of HSA-AR was found at 6 weeks of age. Analyses of body composition

found that HSA-AR increases LBM% (F3,44 = 13.249, P < 0.01) and decreases FBM%

(F3,44 = 14.335, P < 0.001). These effects occur in the same direction as those seen due to

HSA-AR in males. No effects due to Tfm were found, although females can only be

heterozygous for the mutation. As mentioned, these effects of the transgene were

transient, and disappeared when females were again measured at 8 weeks and 10 weeks.

Generally speaking, therefore, differences in body composition due to HSA-AR

expression are not seen in females, likely due to low levels of circulating T.

3.3.5. Individual Muscle and Fat Pad Weights

In order to confirm the differences in body composition seen in HSA-AR males,

adult male rats were dissected (16 weeks of age), and individual skeletal muscles and fat

pads were weighed. Analysis of EDL muscle mass (Fig 4a) shows no effect of HSA-AR

on total mass (F3,45 = 0.650, P = 0.847). Tfm males did have lower EDL mass (F3,45 =

6.083, P < 0.05), although this was expected due to lower overall body weight found in

male mutants. Similarly, mass of individual AT muscles (Fig 4b) also was not affected by

HSA-AR expression (F3,45 = 3.70, P = 0.546). Reduction in AT mass due to Tfm was not

quite statistically significant, although it strongly approached the set α value (F3,45 =

3.874, P = 0.055). Thus, the mass of individually dissected skeletal muscles does not

differ between HSA-AR and WT animals. This confirms the analysis derived from DXA

scanning, which found no differences in raw LBM due to HSA-AR expression.

Page 98: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

88

To confirm differences found in FBM through DXA, visceral WAT fat pads were

dissected from perigonadal depots of male rats and weighed. Analysis of fat pad mass

(Fig 4c) found that these pads are significantly reduced in mass in HSA-AR rats (F3,45 =

16.370, P < 0.001). Thus, transgene expression seems to decrease raw FBM. No effect of

Tfm was found (F3,45 = 2.200, P = 0.147). No interactions were found. Pairwise

comparisons of animals on the same genotypic background showed that HSA-AR reduces

fat pad mass. HSA-AR males have lighter fat pads than WT males (t29 = 1.877, P < 0.05),

and HSA-AR/Tfm males have lighter fat pads than Tfm males (t16 = 3.557, P < 0.05).

Therefore, analysis of individual fat pads confirms the analysis derived from DXA,

suggesting that HSA-AR (or, more generally, myocyte AR) is capable of reducing FBM.

3.3.6. HSA-AR Similarly Affects Body Composition in L78 Mice

We conducted body composition analysis on HSA-AR L78 mice (Monks et al.,

2007) in order to generalize the effects of the transgene in other species, as well as rule

out the possibility of regulation of these effects by ectopic expression of the transgene in

other muscle tissues (such as smooth and cardiac muscles). The HSA-AR mouse lines

have been shown to express the transgene only in skeletal muscle (Monks et al., 2007),

whereas our previously discussed RT-PCR data reveals transgene expression in heart and

smooth muscle of HSA-AR rats. DXA analysis was conducted on WT and L78 HSA-AR

male mice at 24 weeks of age, with representative DXA X-ray images shown (Fig 5a).

Measures of whole body mass (Fig 5b) reveal that L78 mice have lower levels of body

mass than WT littermates (t9 = -5.594, P < 0.001), which is consistent with previous

measures of body mass in this HSA-AR line (Monks et al., 2007). Measures of body

Page 99: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

89

composition indicate striking similarities to the effects seen in HSA-AR rats. L78 males

demonstrate increased LBM% (Fig 5c; t9 = 2.940, P < 0.05) and reduced FBM% (Fig 5d;

t9 = -3.860, P < 0.01). Analysis of raw values indicates that L78 mice have reduced LBM

as compared to WT littermates (Fig 5e; t9 = -2.994, P < 0.05), which is characteristic of

mild muscular atrophy seen in this line (Monks et al., 2007). L78 mice also exhibit

drastically reduced raw FBM (Fig 5f; t9 = -3.860, P < 0.01).

Taken together therefore, analysis of body composition in L78 HSA-AR male

mice reveals effects that largely parallel those seen in HSA-AR rats, including increased

LBM%, and decreased FBM (both raw and when corrected for body mass). This strongly

implicates AR in skeletal muscle myocytes as a major regulator of rodent body

composition.

3.3.7. Effects of HSA-AR and Tfm on Bone Content and Density

Finally, we finished our analysis of body composition in HSA-AR rats by

examining bone mineral content (BMC) and density (BMD). Bone resorption is impaired

in ARKO mice (Callawaert et al., 2009; Kawano et al., 2003), suggesting that AR is

important for maintenance of bone strength. We examined parameters of bone strength in

male and female rats, as well as male L78 mice. BMC refers to the total amount of bone

mass in the body. BMD is measured by correcting BMC for total body volume. However,

because the DXA machine is incapable of determining total body volume, it instead uses

scanned bone area in order to determine BMD values. For that reason, differences in total

body/bone area can strongly skew any differences in BMD.

Page 100: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

90

Unexpectedly, we found that HSA-AR significantly decreases BMC in male rats

(Fig 6a). No differences were found at 4 weeks, but a main effect of HSA-AR is evident

at 6 weeks (F3,42 = 5.659, P < 0.05), 8 weeks (F3,34 = 6.096, P < 0.05), and 10 weeks (F3,34

= 9.542, P < 0.01). Tfm was also found to decrease BMC at these age points as well,

which was to be expected due to lower body mass in the mutant males. When BMD

values were derived for males, however, these effects due to HSA-AR were abolished

(Fig 6c). No differences in BMD were found due to HSA-AR between male rats at 6

weeks (F3,45 = 0.410, P = 0.526), 8 weeks (F3,34 = 0.042, P = 0.839) and 10 weeks (F3,34 =

0.650, P = 0.431). Interestingly, a main effect of Tfm was found in reducing BMD in

males at 4 weeks of age (F3,25 = 5.655, P < 0.05). While this effect is similar to that seen

in male ARKO mice (Kawano et al., 2003), it is transient, and does not persist post-

puberty.

Examination of bone density in females seems to support the results derived from

males. For the most part, no differences are found in BMC in females (Fig 6b). However,

a transient effect of HSA-AR is found at 6 weeks of age, with transgene expression

reducing BMC in females (F3,44 = 10.951, P < 0.01). The directionality of this effect is

consistent with that seen in males, and is also seen transiently at 6 weeks (which is

consistent with other changes in body composition seen in HSA-AR females). The lack of

differences at other age points likely reflects lower circulating T levels in females. No

effects of Tfm are found with regards to BMC. Analysis of BMD in females (Fig 6d)

finds no significant effect of either HSA-AR or Tfm.

Finally, we examined parameters of bone strength and density in HSA-AR L78

male mice. Similar to the effects seen in HSA-AR rats, L78 male mice also have lower

Page 101: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

91

levels of total BMC, as compared to WT littermates (Fig 6e; t9 = 2.298, P < 0.05).

However, as previously mentioned, there is a significant difference in total body mass

between WT and L78 males. Analysis of BMD from these mice reveals no significant

differences between groups (Fig 6f; t9 = 0.249, P = 0.809).

Therefore, in summary, these results indicate that transgene expression results in

reduced BMC in HSA-AR male rats and mice, but not female rats (although this effect

exists transiently at 6 weeks of age). The fact that this effect persists in male mice

suggests that AR in skeletal muscle may possibly be mediating bone development.

Whether this difference in BMC is related to direct action of myocyte AR, or due to

indirect influences (such as differences in FBM), remains to be seen.

3.4. Experiment III: Testosterone Treatment of Adult Female Rats

We found that transgene expression had significant effects on body composition

of male HSA-AR rats and mice, but not in female rats. Thus, we attempted to induce

these previously described effects in adult female rats through acute treatment with

exogenous T. Females had body composition measures completed at baseline. Following

baseline measures (Week 0), they were treated with T capsules for a period of 4 weeks

(Weeks 1-4), during which time they had their body composition analyzed weekly. After

this (at Week 4), T capsules were removed, replaced with vehicle (control) capsules, and

body composition was measured weekly again for 4 weeks (Weeks 5-8). We were mainly

interested in seeing how exogenous T treatment could regulate body composition in HSA-

AR females. Separate ANOVAs were carried out for each phase of the treatment regimen:

the ‘T-Treatment’ phase (Weeks 1-4, using Week 0 as a baseline reference) and the ‘T-

Page 102: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

92

Withdrawal’ phase (Weeks 5-8, using Week 4 as a baseline). ANOVAs examined within-

subject effects on body composition parameters over the treatment, between-subject

effects of HSA-AR, and an interaction between the two. When an interaction was found,

a pair-wise t-test was conducted between the week being measured, and the relative

baseline week (for comprehensive statistical values, see Appendix A). Representative

DXA images of a WT female and her HSA-AR littermate over the course of the treatment

regimen are depicted (Fig 7a).

3.4.1. T-Treatment Regulates Body Composition of HSA-AR Females

We first examined HSA-AR females, in order to determine if their body

composition was significantly affected by treatment with exogenous T.

During the ‘T-Treatment’ phase, LBM% significantly increased in HSA-AR

females, with a main effect of HSA-AR found between-subjects over this treatment

regimen (Fig 7b). HSA-AR females experienced a significant increase in LBM% at 3

weeks (t8 = -3.433, P < 0.01) and 4 weeks (t8 = -2.614, P < 0.05), as compared to baseline

values. HSA-AR females also experienced a significant reduction in FBM% over this

phase of treatment (Fig 7c). A main effect of reduced FBM% was found within-subjects.

Pairwise comparisons show that HSA-AR females had lower FBM% after 3 weeks (t8 =

3.406, P < 0.01) and 4 weeks (t8 = 2.687, P < 0.05) of the ‘T-treatment’ phase, as

compared to baseline values. These differences due to treatment carried over when HSA-

AR animals were examined for changes in raw mass. Unlike HSA-AR males measured

previously, T-treated HSA-AR females experience increases in raw LBM (Fig 7d).

Compared to baseline, HSA-AR females show increases in raw LBM beginning as early

Page 103: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

93

as week 1 (t8 = -3.036, P < 0.05), with consistently elevated LBM at weeks 2 (t8 = -3.972,

P < 0.01), 3 (t8 = -5.617, P < 0.001) and 4 (t8 = -5.064, P < 0.001). Similar changes are

seen in raw FBM of HSA-AR females over the course of T-treatment. Overall, during this

phase, HSA-AR females experience a decrease in raw FBM (Fig 7e). While differences

in LBM are seen rather quickly, reduction in raw FBM is not seen until 3 weeks

following T-treatment (t8 = 2.470, P < 0.05). Thus, it appears that T-treatment of adult

HSA-AR females improves body composition, but that effects in lean muscle tend to

precede those occurring in WAT.

After 4 weeks, T-capsules were removed and replaced with empty vehicle

capsules, followed by weekly body composition measures. During this ‘T-Withdrawal’

phase, HSA-AR females found their body composition parameters returning toward their

original baseline levels. LBM% was significantly lower than week 4 (last week of T-

treatment) by week 6 (t8 = 4.604, P < 0.01), and persisted at week 7 (t8 = 2.337, P < 0.05)

(Fig 7b). Similarly, FBM% was increased in HSA-AR females following T-withdrawal,

with FBM% being significantly higher than week 4 by week 6 (Fig 7c; t8 = -4.724, P <

0.001). Raw values of body composition also seemed to regress back toward original

(week 0) baseline measures following removal of T. The impressive increases in LBM

were followed by a reduction in this parameter by week 6 (Fig 7d; t8 = 5.675, P < 0.001).

FBM, which had been reduced over the course of T-treatment, also increased following

T-withdrawal (Fig 7e). As compared to week 4, FBM values had significantly increased

by week 6 (t8 = -3.720, P < 0.01). This data supports the postulation that the effects of the

transgene are dependent upon circulating T, as subsequent removal of exogenous T

results in abolishment of these effects.

Page 104: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

94

3.4.2. T-Treatment Does Not Affect Body Composition of Wild-type Females

Next, body composition of WT females was examined over the course of the

treatment regimen. Unlike HSA-AR females, it was found that T treatment did not

significantly alter body composition parameters in WT females. No significant effects of

HSA-AR were found between subjects across treatment, nor were any within-subject

differences found for any of the parameters, including LBM% (Fig 7b), FBM% (Fig 7c),

raw LBM (Fig 7f) and raw FBM (Fig 7g). This suggests that the increase in exogenous T

given to WT females had no effect on overall body composition. Thus, this data indicates

that in HSA-AR females, the induction of changes in body composition are due to

transgene expression in myocytes, and not simply due to increased levels of circulating T.

3.5. Experiment IV: Adipose Histology

Previously discussed results indicate that muscle AR is sufficient for the reduction

of adiposity that is seen in HSA-AR rats. This finding was unexpected, and we chose to

examine it further on a cellular level through analysis of individual adipocytes. Thus, at

16 weeks of age, adult male rats were overdosed with sodium pentobarbital and

perigonadal fat pads were dissected for adipose histology.

3.5.1. HSA-AR Expression Reduces Adipocyte Area

Representative images of adipocytes from WT, HSA-AR, Tfm, and HSA-AR/Tfm

adult male rats are shown (Fig 8a). We measured adipocyte area in order to determine if

the size of these cells was altered by transgene expression. Interestingly, it was found that

Page 105: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

95

HSA-AR expression reduces overall adipocyte area (Fig 8b). A main effect of HSA-AR

was found, with male HSA-AR animals demonstrating reduced adipocyte area, as

compared to WT littermates (F3,18 = 16.370, P < 0.001). No main effect of Tfm was found,

although Tfm males (without the transgene) were found to have the largest adipocyte area,

when compared to WT animals, which had the next highest area (t10 = -2.645, P < 0.05).

No interaction was found between HSA-AR and Tfm.

Frequency distributions were then calculated, in order to visually depict

differences in proportions of cell size (Fig 8c). As can be seen, male rats expressing the

transgene experience a shift toward the left (smaller cells), as compared to WT littermates

on the same genetic background. This indicates that HSA-AR males have an increased

proportion of smaller adipocytes than WT males. Taken together, therefore, these results

indicate that differences in adiposity between WT and HSA-AR males can be

demonstrated at a cellular level, with reduced size of individual adipocytes noted in HSA-

AR animals.

3.6. Experiment V: Energy Balance and Metabolic Analyses

We hypothesized that the differences seen in body composition of HSA-AR males

(particularly the reduction of FBM) were mediated by changes in overall energy balance

and metabolism. Reduction in overall FBM may occur due to increases in energy

expenditure. Thus, we examined energy expenditure in HSA-AR animals by examining

basal metabolism (using the indirect measure of O2 consumption) and spontaneous

activity level.

Page 106: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

96

3.6.1. HSA-AR Expression Increases Resting Metabolism in Rats and Mice

Analysis of resting metabolic rate (RMR) was accomplished through the use of

indirect calorimetry. In this case, O2 consumption was measured, with higher levels of

this parameter associated with increased RMR (Holloszy, 2008). At 12 weeks of age,

male rats were placed in a gas-exchange chamber, and O2 consumption was measured

through the use of an O2 sensor (Fig 9a). Interestingly, a main effect of HSA-AR was

found, with transgene expression being associated with higher levels of O2 uptake (F3,35 =

10.817, P < 0.01). No main effects for Tfm, or an HSA-AR X Tfm interaction, were

found. However, when RMR was derived after correction for body weight (Fig 9b), a

main effect of HSA-AR was not noted, although it did approach significance (F3,35 =

7.574, P = 0.1). Here Tfm actually demonstrated a significant effect (F3,35 = 14.348, P <

0.001), which was to be expected due to the lower body mass of Tfm males. An

interaction between HSA-AR and Tfm was found (F3,35 = 1.40, P < 0.01), with HSA-

AR/Tfm males demonstrating the highest RMR levels.

We also examined O2 consumption and RMR in HSA-AR L78 mice, in order to

test the generality of our effects (Fig 10a). WT and HSA-AR male mice were measured in

adulthood (approximately 24-30 weeks of age), using a similar paradigm as that which

was used for rats. Although there was a trend toward higher O2 consumption in HSA-AR

L78 male mice (as compared to WT littermates), this effect was not significant (t9 = -

0.828, P = 0.429). However, derivation of RMR (which accounts for disparities in overall

body mass), indicates that RMR levels are increased in HSA-AR L78 males (Fig 10b; t9 =

-2.286, P < 0.05). Therefore, this evidence seems to indicate that HSA-AR male rodents

Page 107: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

97

may be hypermetabolic, resulting in increased rate of energy expenditure, and

subsequently providing a mechanism for the reduction in adiposity seen in these animals.

3.6.2. Spontaneous Activity is Not Affected by the Transgene

Another major component of energy expenditure is physical activity, and thus we

examined this parameter in HSA-AR male rats. ARKO mice have been shown to

demonstrate reduced voluntary activity, suggesting that AR-activation may be important

in mediating this behaviour (Ophoff, Callewaert et al., 2009). Animals were investigated

at the same ages at which body composition was analyzed (4, 6, 8 and 10 weeks of age).

No differences in spontaneous activity were found between groups at any age of

examination (Fig 11). No main effects of HSA-AR or Tfm (or interaction between the

two) were found for male rats at any time-point in question. Thus, it remains unlikely that

AR in myocytes is capable of modulating physical activity in these animals.

3.7. Summary

Characterization of HSA-AR expression in transgenic animals shows that the

transgene is expressed only in myocytes. Expression of the transgene is associated with

profound changes in body composition of HSA-AR male rats and mice, which includes:

Increased LBM%, decreased FBM%, decreased raw FBM, and reduced BMC. These

differences are confirmed by dissection and weighing of individual muscles and fat pads.

All of these findings are also demonstrated in HSA-AR/Tfm rats, suggesting that AR in

myocytes alone is sufficient to stimulate these effects. While these changes in body

composition are not found in HSA-AR female rats, they can be induced through acute T

Page 108: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

98

treatment, showing that these effects are likely T-dependent, and do not occur as a

consequence of transgene expression alone. Finally, it has been shown that HSA-AR

expression modulates metabolic parameters, inducing energy expenditure by elevating

RMR, but not affecting activity level. Please refer to Appendix A for full statistical

analyses.

Page 109: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

99

Chapter 4: Discussion 4.1. Overview

Taken together, our results present an in vivo model whereby increased androgen

signaling in myocytes alone is sufficient to induce robust changes in body composition

(increasing LBM% and reducing FBM%) and systemic metabolism. These differences

were also found in HSA-AR L78 male mice, indicating that they are a general result of

overexpression of AR in skeletal muscle myocytes. In addition, several of our results

suggest that these effects are dependent upon T. First, in males, the effects were not

evident until 6 weeks of age, when these animals reach puberty and circulating T levels

increase drastically. Secondly, in female rats, these effects could be induced through

acute treatment with exogenous T, indicating that transgene expression alone was not

sufficient (although one cannot rule out the potential roles of female sex chromosomes

and ovarian hormones). Finally, in these T-treated female rats, removal of T abolished

these effects, and females returned to their baseline levels of body composition within 1-2

weeks. Therefore, it is clear that the differences induced in body composition by myocyte

AR are dependent upon activation of the nuclear receptor by T in this cellular population.

It seems likely that these differences in body composition result from regulation of

systemic metabolism, evidenced by increased oxygen consumption and RMR in HSA-AR

male rats and mice. Finally, no differences were found in spontaneous activity level of

rats, indicating that the augmented RMR level of these animals is likely the driving force

behind their increased energy expenditure and changes in body composition.

Page 110: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

100

4.2. Myocyte AR and Body Composition

This set of experiments provides insight into how androgens affect body

composition. Notably, they provide evidence for androgenic action on muscle to reduce

fat via increases in metabolic rate. Despite widespread belief that androgen

supplementation alters body composition, experimental support for this idea has only

recently been obtained (Bhasin et al., 1997). Furthermore, T-treatment of older men has

been successful in limiting the negative effects of aging on body composition, with some

scientists citing the reduced T levels that men experience as they age as a prominent

contributing factor to late-onset metabolic disease (Bhasin et al., 2006). These effects of

androgens on body composition have been verified by basic science, gene-targeting

research on ARKO mice. Ablation of AR throughout the body results in atrophy of

skeletal muscle (MacLean et al., 2008), adult-onset obesity (Lin et al., 2005; Fan et al.,

2005) and hypometabolism (Fan et al., 2005). While these effects are interesting, and

provide further credence to the idea that androgens are capable of modulating body

composition, they do not identify the tissue-specific AR that are necessary for mediating

them. Identifying these targets is a major goal of research in androgen therapy, for the

purposes of exploiting them as treatments for obesity and metabolic disease. Our study

employed a gain-of-function transgene (HSA-AR) that results in overexpression of AR

protein in myocytes. Furthermore, by crossing HSA-AR rats with those possessing the

Tfm mutation, we studied the resultant HSA-AR/Tfm progeny, which express AR only in

myocytes. Using these powerful genetic tools, we have gained significant insight into

how AR in myocytes contributes to changes in body composition.

Page 111: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

101

4.2.1. Lean Muscle Mass

In popular culture, the association between androgens and skeletal muscle is a

common one (Kadi, 2008). The use of anabolic-androgenic steroids (which are

pharmacological agents analogous in structure to endogenous androgens, and that also

bind to AR) has been linked with performance-enhancement in various sports – with the

belief that use of these drugs leads to increased lean muscle mass, and thus increased

strength and power. More specifically, human studies in adult men have found that T

supplementation has widely been associated with hypertrophy of skeletal muscle fibers

(Sinha-Hikim et al., 2002). T-dependent hypertrophy has been found in young men, as

well as through T-treatment of elderly males (Bhasin et al., 2001). Androgens have found

limited clinical use, but have been shown to have beneficial effects in increasing muscle

mass of patients suffering from HIV wasting (Bhasin et al., 2000), renal disease

(MacDonald et al., 2007) and burns (Wolf et al., 2006). However, despite these

associative clinical studies, the idea that T and other androgens directly increase lean

muscle mass is not completely accepted, due to limited evidence of sufficient functional

benefits in T-treated humans (MacLean & Handelsman, 2009).

For this reason, basic science research using cell cultures and animal models have

attempted to bridge this gap of knowledge by identifying cellular and molecular

mechanisms by which androgens/AR can influence lean muscle mass. Developmental

work has shown direct function for T in progenitor cells. Treatment of pluripotent stem

cells (or more mature satellite cells) with T enhances commitment of these cells to the

myogenic lineage, proliferation of satellite cells, and myoblast/myofiber protein accretion

(Singh et al., 2003; Joubert & Tobin, 1995; Chen, Lee, Zajac & MacLean, 2008).

Page 112: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

102

Treatment of these cells with an AR antagonist (such as flutamide) abolishes these

effects. Therefore, it is largely well-accepted that androgens have developmental effects

in promoting myogenesis. However, identifying mechanisms by which androgens can

contribute to hypertrophy of mature skeletal muscle remains elusive.

Work using animal models has provided some promising evidence of AR effects

on adult muscle. Abolishment of AR in ARKO mice results in reduced muscular strength

and muscular atrophy (MacLean et al., 2008). However, the nature of this knockout

means that AR has been deleted from all cells, including important myocyte precursor

cells. Thus, teasing out the effects of AR in myogenesis from those in adult muscle using

whole-body ARKO is not feasible. To get around this problem, Ophoff and colleagues

(2009) used the Cre/Lox approach to generate myocyte-specific ARKO mice (mARKO).

This was done using the myocyte-creatine kinase (MCK) promoter to drive Cre-

expression, so that AR is deleted only in mature myocytes that express this promoter, but

not precursor cells (such as satellite or embryonic stem cells). The muscle phenotype in

mARKO mice is very distinct, with KO mice demonstrating reduced limb muscle mass

and strength. These mARKO mice also exhibit a change in fiber type of soleus muscle,

with an increase in type I fibers. It is also worth noting that the atrophy found in skeletal

muscle of mARKO mice is not as severe as that seen in whole-body ARKO mice,

implying the importance of AR in progenitor cells, as well as the possibility of

involvement of AR in other tissues.

In contrast to the loss-of-function paradigm employed by Ophoff et al., we used a

gain-of-function transgenic model to overexpress AR in skeletal muscle fibers. This was

done through the use of the HSA promoter. We find that overexpression of AR in adult

Page 113: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

103

muscle fibers increases LBM%, but that these effects do not carry over when

investigating raw values in males. That is, adult HSA-AR males (6 to 10 weeks) do not

demonstrate increases in raw LBM. This lack of difference was confirmed through

dissection of individual muscles, as EDL and AT muscle weight did not differ between

WT and HSA-AR littermates. However, T-treatment of adult HSA-AR female rats

resulted in significant increases in LBM, effects that were not seen through T-treatment of

WT females. Furthermore, removal of T caused a significant decline in LBM. Thus, it is

possible that myocyte AR have acute, local functions in increasing skeletal muscle mass.

Histological analysis of skeletal muscle of HSA-AR male rats by our lab also

reveals interesting insights (Fernando et al., 2010). Dissected EDL muscles from intact

HSA-AR and WT rats were obtained at 10 weeks of age. Analysis of number of

myofibers revealed no differences between WT and HSA-AR animals. Furthermore,

fiber-type analysis shows no shift in the fiber-type proportion. Interestingly, however,

selective hypertrophy of type IIb (fast-twitch, glycolytic) muscle fibers was noted. These

muscle fibers showed larger area, while other fiber-types were not significantly different

between groups. Ophoff et al. (2009) found that myocyte-specific ARKO also had no

effect on fiber-type proportion in EDL, although they did not report any differences in

fiber hypertrophy either.

On the face of it, our findings seem largely incongruent to those reported by

Ophoff et al. in their mARKO mouse model (2009). While these scientists report that

knockout of AR in myocytes reduces muscle mass, we found no effect on raw muscle

mass through overexpression of AR in myocytes (particularly in the EDL). However,

very recent evidence from Chambon and colleagues (2010) seems to support our findings.

Page 114: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

104

Here, scientists developed another mARKO model through the use of the HSA promoter.

In this study, it was found that (in contrast to Ophoff et al.) selective ablation of AR in

myocytes did not affect limb muscle mass (including the EDL), but rather had significant

effects on the strength of these muscles. Furthermore, the authors also show reduced

cross-sectional area of type II muscle fibers in the EDL, which nicely compliments the

selective hypertrophy of these fibers seen in our transgenic rats. Thus, our HSA-AR

model seems to indicate that androgens may have effects on mature myocytes. However,

the nature of these effects remains unclear. Whether myocyte AR plays a significant role

in increasing muscle mass in non-sexually dimorphic muscles will continue to be a hotly

debated topic.

4.2.2. Adipose Tissue

While research into androgen effects on skeletal muscle mass have resulted in

controversial results, very little work has been undertaken in order to determine the nature

of how androgens affect adipose tissue. Clinical studies on patient populations show that

exogenous T treatment can have beneficial effects in reducing adiposity in young and

older men (Bhasin et al., 1996; Bhasin et al., 2001). In addition, epidemiological surveys

reveal that serum T levels are inversely related to whole-body and regional fat mass in

men (Siedell et al., 1990; Derby et al., 2006). However, identification of the cellular and

molecular mechanisms involved in this androgenic reduction of fat has not been well

elucidated.

In the previous section, it was mentioned that androgens are believed to have dual

effects on skeletal muscle, mediating the development of mature myocytes from

Page 115: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

105

progenitor cells, as well as hypertrophy of existing, mature cells. However, in study of

adipose tissue, androgens (such as T) have been implicated largely in inhibiting the

development of mature adipocytes from precursor cells (termed ‘pre-adipocytes’). This

process by which progenitor precursor cells develop into mature adipocytes is known as

adipogenesis. While originally believed to be a process that occurred only during the

early stages of life, it is now known that adipogenesis occurs across the lifespan (Rosen &

Spiegelman, 2000). For this reason, reduction of adiposity (lower fat accumulation) in

adulthood can either be the result of increased fatty acid β-oxidation (metabolism of fat)

or inhibition of adipogenesis. As mentioned, androgens have traditionally been associated

with the latter process. Significant in vitro work using human and rat pre-adipocytes has

shown that differentiation of pre-adipocytes into mature adipocytes is impaired when

cells are treated with T (Singh et al., 2003; Singh et al., 2006; Gupta et al., 2008). Gene

expression assays of these cells reveal significant down-regulation of major adipogenic

factors, such as PPARγ and C/EBPα. Furthermore, this androgenic regulation of

adipogenesis is believed to be mediated by activation of AR, as pre-treatment of cells

with an AR antagonist abolishes these effects. Thus, it has largely been believed that

androgens exercise their effects on adipose tissue by binding to AR in adipocytes, and

subsequently down-regulating major adipogenic genes.

Manipulation of AR using in vivo animal models has been largely consistent with

the idea of androgen-mediated reduction in adipose tissue. Whole-body ablation of AR

results in adult-onset obesity, with significantly increased adipocyte size, and higher

frequency of larger adipocytes in ARKO mice (Lin et al., 2005; Fan et al., 2005).

Interestingly, ARKO mice were shown to have reduced expression in adipose tissue of

Page 116: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

106

adipogenic genes (including PPARγ and C/EBPα), but also decreased expression of

major genes in liver and skeletal muscle that are involved in β-oxidation (such as

PPARα). Thus, it was unclear as to whether the increased adiposity seen in late-adulthood

of ARKO mice was due to reduced adipogenesis or reduced fatty acid metabolism (or

both). Also, although these findings were interesting, they did not shed any light on which

tissue-specific AR were responsible for the increase in WAT seen in ARKO mice. With

this in mind, scientists generated mice with selective knockout of AR in adipocytes

(aARKO; Yu et al., 2008), since (as mentioned) it was largely believed that AR in these

cells mediate T’s effects on WAT by inhibiting adipogenesis. Unexpectedly, these

aARKO mice did not demonstrate the adult-onset obesity seen in whole-body ARKO.

These same scientists then examined selective knockout of AR in liver hepatocytes

(hARKO), a major organ involved in fat catabolism (Lin et al., 2008). While hARKO

mice demonstrate reduced hepatic PPARα expression, and hepatic steatosis (local

accumulation of fat in the liver), they also do not show adult-onset obesity typical of

ARKO mice. Thus, it appears that neither AR in adipocytes nor hepatocytes are sufficient

for regulation of the androgenic reduction in adiposity.

We examined FBM in our HSA-AR rats and mice, which overexpress AR in

skeletal muscle myocytes. Skeletal muscle is an attractive but often overlooked tissue in

the study of regulation of FBM. Myocytes are energetically-demanding cell populations,

and disruption of fatty acid transport into myocytes is often associated with metabolic

diseases (Zitzmann, 2009). We found that FBM is drastically reduced in HSA-AR rats

and mice, a finding that is confirmed through dissection and weighing of individual fat

pads. In rats, these effects are seen shortly after the onset of T secretion during puberty,

Page 117: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

107

suggesting that they are T-dependent. Examination of L78 mice in late adulthood shows

that this effect persists. Furthermore, T-treatment of HSA-AR female rats reduces

adiposity significantly – an effect which is abolished upon withdrawal of T. Examination

of adipocyte size in HSA-AR males reveals that they have smaller cell area than WT

littermates, and an increased proportion of these smaller cells.

These findings, that HSA-AR expression reduces fat mass and adipocyte size,

were largely surprising because AR overexpression is limited to myocytes (in L78 mice,

only in skeletal muscle myocytes). Furthermore, they were also seen in HSA-AR/Tfm

rats, which express AR only in myocytes, as provided by the transgene. This suggests that

myocyte AR is sufficient to reduce FBM. These effects are in contrast to the more

traditional model of androgens reducing WAT through activation of AR in adipocytes,

and subsequent inhibition of adipogenesis. While our model was not designed to test the

function of AR in adipocytes, it can be said with some certainty that action of AR in

WAT is not responsible for the results that we see. This is for several reasons. First, HSA-

AR mRNA expression was not found in WAT of rats through RT-PCR. Examination of

β-galactosidase expression in L78 mice also found no expression of AR in this cellular

population (Monks et al., 2007). Furthermore, these effects were also seen in HSA-

AR/Tfm male rats, which do not express AR in any non-muscle tissue, including WAT

tissue. Finally, adipogenesis is a process that is largely mediated by actions in WAT

tissue itself (Rosen & Spiegelman, 2000). It is therefore highly unlikely that local actions

of AR in muscle could be capable of inhibiting adipogenesis. Given this unlikely

scenario, logic therefore suggests that myocyte AR reduces adiposity by increasing fatty-

acid metabolism (i.e. β-oxidation). This would explain why knockout of AR only in

Page 118: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

108

adipocytes (Yu et al., 2008) or hepatocytes (Lin et al., 2008) is incapable of recapitulating

the adult-onset obesity that is seen in whole-body ARKO mice (Lin et al., 2005). It would

also explain the hypometabolism that is found in whole-body ARKO mice (Fan et al.,

2005), a finding that will be elaborated upon in a later section.

4.2.3. Bone

Sex hormones are well-known to be mediators of bone development and

remodeling during adulthood (Bland, 2000; Compston, 2001). The most prominent

effects of sex hormones in this context are often attributed to estrogens, which directly act

upon ER in bone tissue in order to increase bone strength and remodeling (Simpson &

Davis, 2000; Couse & Korach, 1999). This explains the significant increase in incidence

of osteoporosis after menopause in women, when secretion of estrogens declines

significantly – an effect that can be reduced with treatment of exogenous estrogens

(Riggs, Khosla & Melton, 2002).

The role of androgens in bone development and remodeling, however, is less

understood. AR is expressed in bone tissues, including osteoblasts and osteoclasts

(Compston, 2001). Furthermore, androgens have been shown to modulate expression of

various growth factors and cytokines involved in bone remodeling (Compston, 2001). For

example, T treatment of an osteoblastic cell line increases expression of Insulin-like

growth factor 1 (IGF-1), which is a prominent growth factor that promotes bone

development (Gori, Hofbauer, Conover & Khosla, 1999). Taken as a whole, most

associative research seems to indicate that androgens have protective functions on bone,

resulting in increased bone mass and density in males and females (Bilezikian, 2002;

Page 119: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

109

Hofbauer & Khosla, 1999). Investigation of bone parameters in whole-body ARKO mice

seems to support these findings (Kawano et al., 2003). 8-week old male ARKO mice

demonstrate high bone turnover with increased bone resorption. Overall bone mineral

content (BMC) values are reduced in these ARKO mice, and analysis of primary

osteoblast/osteoclast cultures from ARKO mice reveals that reduced androgen signaling

removes inhibition of osteoclastogenesis, resulting in breakdown and resorption of bone.

Therefore, androgens are believed to be major regulators of bone development and

remodeling, and act by binding to AR in bone and directly inhibiting osteoclastogenesis.

We investigated both BMC and bone mineral density (BMD) in HSA-AR male

rats and mice. Surprisingly, we found that BMC was significantly reduced in male HSA-

AR animals, both in rats and mice. This effect was also not present at 4 weeks of age, but

emerged after the onset of puberty, suggesting that it is T-dependent. Furthermore, this

effect was seen transiently at 6 weeks in HSA-AR female rats, a time period where other

changes in body composition were found (LBM% and FBM%). Reduced BMC is also

seen in HSA-AR/Tfm males, as compared to Tfm males, suggesting that AR in myocytes

alone is sufficient for this reduction in bone mass. No effects of HSA-AR were found on

BMD, however this measure may be confounded, as it is incorrectly calculated by the

DXA machine using bone area and not total volume. Interestingly, no effects of Tfm were

found with regards to BMD, although at 4 weeks, male rats possessing this mutation

exhibited lower BMD as compared to WT controls.

Our findings lie in stark contrast to the effects found in ARKO mice (Kawano et

al., 2003), and the typically beneficial effects on bone attributed to androgens (Bilezikian,

2002). While these studies suggest that androgens are important in maintaining and

Page 120: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

110

increasing bone mass, we find a reduction of BMC in animals overexpressing AR in

myocytes, and those expressing AR only in myocytes. These results suggest two possible

explanations. First, it is possible that action of AR directly in myocytes is contributing to

reduction of BMC. Although we did not examine expression of various bone growth

factors and cytokines, it is unlikely that myocyte AR down-regulates these factors. In

actuality, existing lines of evidence seem to indicate the opposite: that AR activation in

muscle actually promotes increases in expression of IGF-1 signaling molecules, although

whether these molecules are then capable of circulating throughout the body is unknown

(Svensson et al., 2010). A more likely explanation for the reduction in BMC seen in

HSA-AR animals is that it is due to indirect effects of the transgene – namely the

reduction in adipose tissue. Increases in WAT are paradoxically known to be associated

with decreased serum levels of adiponectin, an adipocyte-derived cytokine, which is

sometimes classified as a hormone (Arita et al., 1999). Very recent work examining

adiponectin-knockout mice reveals that these animals have higher levels of BMC, and

lower bone fragility (Williams et al., 2009). While we did not examine adiponectin

concentrations in our animals, it is reasonable to assume that WT animals would have

lower levels of the hormone, due to increased FBM. Lower levels of adiponectin would

thus explain the increase in BMC seen in WT animals, similar to those seen in

adiponectin-knockout mice. Nevertheless, more research is necessary in order to

determine mechanisms of muscle-bone and WAT-bone crosstalk.

Page 121: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

111

4.3. The Effect of Tfm on Body Composition

Our work also allows for the understanding of how whole-body ablation of AR

can contribute to changes in body composition, through the use of male Tfm rats. To that

end, we find several interesting differences in body composition between WT and Tfm

male rats that are worth discussing.

4.3.1. Comparison with ARKO Mice

As mentioned previously, several distinct phenotypes are found with regards to

body composition in ARKO mice (as compared to WT littermates). In the context of

skeletal muscle, ARKO males demonstrate muscular atrophy as well as reduced muscle

mass, coupled with impaired force production (MacLean et al., 2008). These findings also

extend to cardiac muscle, with reduced heart mass and atrophy of cardiomyocytes in

ARKO males (Ikeda et al., 2005). FBM also appears to differ in ARKO mice, as male

ARKO demonstrate adult-onset obesity (development of excess adiposity after 30 weeks

of age), coupled with increased size of individual adipocytes and heavier fat pads (Lin et

al., 2005; Fan et al., 2005). Lastly, differences in bone were also found in ARKO mice

(Kawano et al., 2003). Male ARKO mice show reduced bone mass, increased resorption

of bone (due to higher rates of osteoclastogenesis) and osteopenia. Taken together, we

can identify a body composition phenotype of ARKO mice characterized by reduced

LBM and BMC, but increased FBM. These findings were only found in males of this line,

and not females.

Our examination of body composition in Tfm males reveals several striking

differences between these mutants and their WT littermates. Examination of LBM per

Page 122: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

112

unit of body mass reveals higher levels of LBM% in Tfm males. With regards to WAT,

we also find that Tfm males demonstrate reduced FBM%, as compared to WT littermates.

However, examination of adipocytes of Tfm males in late adulthood reveals that cells

from these animals have the largest area. Finally, BMD was examined in Tfm males. It

was found that the mutation resulted in reduced BMD at 4 weeks of age, but this effect

was transient, and not found in examination of rats at later ages. In summary, therefore,

we find that body composition of Tfm males is marked by increased LBM%, reduced

FBM% and no effects on BMD, as compared to WT littermates. These differences are

largely contradictory to those previously described to have been found in ARKO male

mice.

4.3.2. Why the Discrepancy?

While it is tempting to directly compare Tfm rats with ARKO mice, there are

several important phenotypic discrepancies between the males of these strains (aside from

the obvious species difference) that should be noted. First, the ARKO is derived using

Cre/Lox technology, and subsequent deletion of the AR gene (Yeh et al., 2002). Thus, in

theory, no AR protein can be translated in any cells where this gene has been deleted. In

practice however, the efficacy of Cre-induced recombination in ARKO mice has been

called into question, as the degree of Cre-mediated excision is unknown (MacLean &

Handelsman, 2009). For example, protein expression assays of moderate sensitivity

demonstrate some AR expression in muscle of mARKO mice, although the levels are

significantly lower than those seen in WT controls (Ophoff et al., 2009). Thus, the

potential for residual AR activation remains. In contrast to the Cre/Lox technology used

Page 123: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

113

to generate ARKO, Tfm results from a single base-pair mutation, resulting in the

alteration of a single amino acid in the AR polypeptide (Yarbrough et al., 1990). The

amino acid alteration occurs in the ligand-binding domain of the AR protein, and though

the protein is translated, it is incapable of binding to AR, and thus being activated.

Nevertheless, Tfm AR protein can be identified using immunohistochemistry (IHC), and

some residual AR activation is found in tissues of Tfm males (Zuloaga et al., 2008).

Therefore, there are significant differences between ARKO mice and Tfm rats in terms of

AR protein translation and function.

Another major difference between ARKO mice and Tfm rats is exemplified by

circulating hormone levels. In ARKO mice, serum T and DHT levels are drastically

reduced, as compared to WT males (Yeh et al., 2002; Kawano et al., 2003). This is

believed to be due to atrophy of testes in these animals. Similar findings are found in Tfm

mice (Jones et al., 2003), who demonstrate decreased circulating T levels, due to atrophy

of testes. In contrast, Tfm male rats have been found to have significantly higher levels of

circulating T, reaching the highest limits seen in WT rats (Naess et al., 1976). This occurs

despite testicular atrophy in these animals, and is believed to be due to lack of AR in the

hypothalamus, and thus uncoupling of negative feedback in these animals. This is further

evidenced by increases in gonadotropin (FSH and LH) levels in these rats. Therefore, not

only do ARKO mice and Tfm rats differ in translation of AR protein, but their hormonal

profiles are also rather distinct.

Our findings regarding body composition in Tfm rats are largely contradictory to

the existing literature regarding androgens and body composition (Zitzmann, 2009). First,

it is important to note that these findings in Tfm rats do not necessarily cast doubt on our

Page 124: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

114

findings regarding the HSA-AR transgene and myocyte AR. While Tfm does improve

body composition, rescue of AR in myocytes of Tfm males shifts them further in the same

direction. Therefore, it is unlikely that loss of AR in myocytes of Tfm males causes the

improvement in body composition. Nevertheless, it is puzzling that inactive AR in any

tissue would be capable of improving these parameters.

Various explanations may account for these counterintuitive effects of Tfm. First,

it is important to note the differences in developmental periods at which ARKO mice

were measured, as compared to our experiments in Tfm rats. For example, in the original

study employing ARKO mice, differences in FBM were investigated at 8 weeks of age

(Yeh et al., 2002). Here, the scientists found reduced adiposity and smaller adipocytes in

ARKO mice (similar to our findings in Tfm rats), and initially concluded that AR might

be necessary for adipogenesis. It was not until these animals were subsequently followed

into late-adulthood (30 weeks of age) that increased adiposity was found (Lin et al.,

2005). We investigated Tfm males from only 4 weeks of age until 10 weeks, so it is

possible that these mutants may develop obesity in later adulthood.

The higher levels of circulating T also complicate the picture of Tfm males.

Aromatization of T results in synthesis of estrogens, which have diverse functions (the

possible role of estrogens in mediating differences in LBM and FBM that we see will be

discussed in the next section). Although activity of aromatase enzymes is shown to be

reduced in brain regions of Tfm males (Roselli, Salisbury & Resko, 1987), higher serum

concentrations of estrogens have also been reported in these mutant males, presumably

due to increased levels of T substrate (Vanderschueren, Boonen & Bouillon, 1998). While

we did not measure levels of serum estrogens in our animals, it is possible that elevated

Page 125: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

115

levels would contribute to major differences between Tfm and WT brothers, as estrogens

have major regulatory effects on body composition (namely through reduction of adipose

tissue).

Study of bone structure in Tfm males shows that these animals are able to obtain

similar trabecular bone volumes as WT male littermates (Vanderschueren et al., 1993).

Furthermore, gonadectomy of Tfm males significantly reduces BMD in these animals

(Vanderschueren et al., 1994). This is strong evidence that this effect in Tfm males is a

result of T levels. However, since AR is non-functional in these animals, a metabolite of

T is likely mediating this effect. The authors hypothesize that the metabolite in question

may be estrogen. We have previously discussed that estrogens have beneficial effects in

improving BMC and BMD (Simpson & Davis, 2000). Treatment of male rats with an

aromatase inhibitor (which prevents the conversion of T to estrogens) during

development impairs BMD, highlighting the importance of estrogens in mediating BMD

in both males and females (Vanderschueren et al., 1997). Thus, our experiments show

that Tfm male rats do not display the impaired BMD that is characteristic of ARKO male

mice. However, this protection against osteopenia in Tfm rats (as evidenced by our

results) may potentially be a result of increased estrogen action in these rats; a

consequence of high T levels.

4.4. Interactions with Estrogens

The influence of estrogens is an important aspect that must be discussed, as

estrogens are known to have profound effects upon body composition in both males and

females. These effects have significance not only for the previously mentioned Tfm male

Page 126: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

116

rats (who demonstrate increased estrogen levels), but also for our T-treated adult females,

whose estrogen signaling was likely impaired due to treatment with exogenous T. The

withdrawal of T from these rats originally results in regression toward baseline values

(that is, increased FBM% and decreased LBM%). However, toward the end of

monitoring, both groups display non-significant trends in body composition that mimic

those that occur due to transgene expression (decreased FBM% and increased LBM%).

These latter changes may reflect an increase in estrogenic signaling in females of both

genotypes, due to removal of T, and subsequent increase in circulating gonadotropins.

Thus, understanding of any estrogenic influences on effects of the transgene are important

for fully elucidating the sub-cellular mechanisms that lead to the phenotypes seen.

4.4.1. Estrogens and Adipose Tissue

It has been long understood that estrogen signaling is important in reducing fat

mass in females (Shi et al., 2009). Estrogen receptor null (ERKO) mice demonstrate

increased adiposity as early as 6 weeks of age (Heine et al., 2000). Furthermore, these

effects are apparent in male mice as well as female mice, a finding that is not replicated in

ARKO mice, whose effects are only seen in males (Lin et al., 2005). Thus, estrogens

appear to be important in mediating adiposity in both sexes, although the effects are more

pronounced in females (Heine et al., 2000). In fact, many have made the argument that

androgens exert their effects on body composition in males only indirectly – through

aromatization to estrogens. Aromatase-knockout (ArKO) male mice exhibit obesity in

adulthood, despite elevated serum T levels, and this effect was abolished through

treatment with exogenous 17β-estradiol (Jones et al., 2000). Furthermore, treatment of

Page 127: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

117

gonadectomized male mice in adulthood with DHT (a non-aromatizable androgen) results

in increased fat mass, as compared to a control group treated with 17β-estradiol

(Moverare-Skrtic et al., 2006). Strangely, however, the DHT treated group did not differ

from sham-operated animals. Therefore, it is clear that estrogens have significant effects

on WAT, and many have argued that these effects are more prominent than (or are the

consequence of) effects of androgens.

4.4.2. Estrogens, HSA-AR and Tfm

Given these robust effects of estrogens, it is important to dissect any influence

they may have on the effects that we see. There are two separate venues from our

experiments where estrogens may be especially important. First, as mentioned, Tfm male

rats have been shown to have higher levels of estrogens (Vanderschueren, Boonen &

Bouillon, 1998), which may explain their surprising differences in body composition.

Secondly, we treated WT and HSA-AR adult female rats with exogenous T, in order to

determine the necessity of androgens in mediating this effect. In doing this, estrogen

signaling was likely attenuated significantly, complicating the hormonal state of these

animals.

Before considering the role of estrogens in each of these separate contexts, one

must first confront the mechanism of estrogenic effects on WAT. Interestingly, ERKO

mice do not exhibit significant differences in energy intake, suggesting that this parameter

does not account for the increased adiposity seen in these animals (Heine et al., 2000).

While feeding behaviour was not different between genotypes, energy expenditure (in the

form of O2 consumption) was found to be reduced in male and female ERKO mice.

Page 128: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

118

However, the authors did not disclose whether these differences were due to alterations in

BMR, adaptive thermogenesis or activity level (or some combination of these factors).

Furthermore, it was also unclear as to which tissues were involved in mediating this

estrogenic reduction in adiposity. An answer to both of these questions was provided by

Musatov et al. (2007), who delivered an ER-specific siRNA directly to the ventromedial

hypothalamus, reduced ER expression in this brain area, and found that it resulted in

obesity due to reduced activity levels in treated animals. This work was supported by

research on aromatase-null mice, which also exhibit normal RMR and feeding, but

reduced spontaneous activity (Jones et al., 2000). Despite the evidence cited in these

papers, relatively new work indicates that estrogen signaling in the hypothalamus does

seem to influence feeding behaviour (Shi et al., 2009), and is believed to function by

heightening anorexigenic signaling molecules of the leptin pathway (Gao et al., 2007).

Thus, it appears that estrogens influence body composition (more specifically WAT)

through direct actions on the hypothalamus, and by modulating both feeding behaviour

and activity level.

With this information in mind, one can postulate as to whether estrogen plays a

role in mediating effects that we see due to HSA-AR. First, it is highly unlikely that

estrogen plays a major role in regulating the differences seen in WAT in HSA-AR males.

No differences were found in spontaneous activity of HSA-AR males. Earlier work

monitoring feeding behaviour also shows no significant effect of transgene expression on

food intake (Rao & Monks, unpublished data). Thus, there is little reason to believe that

the effects seen in HSA-AR males can be attributed to influence of estrogen.

Page 129: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

119

Conversely, with regards to the T-treated females, estrogenic effects become a

real possibility, since we did not ovariectomize these females. Thus, one might argue that

the effects seen due to T-treatment might actually be secondary effects caused by

suppression of ovarian hormones. While we did not measure energy intake or expenditure

in these females, this possibility is also unlikely. First, as has been discussed, estrogen

signaling is important for reduction of WAT, but we see a similar result from suppression

of estrogen signaling, suggesting minimal effect of ovarian hormones. Secondly, these

effects manifest themselves only in HSA-AR females, indicating that the dosage of

exogenous T itself (and subsequent suppression of estrogens) was not responsible for

these effects, but rather binding of this T to the transgenic AR. Thus, while estrogens are

important in mediating WAT in females, it is unlikely that they are involved in regulating

the effects that we see in our T-treated females.

Finally, the influence of estrogens in regulating WAT in Tfm males is possibly a

very real one. It has been shown that estrogen levels are significantly elevated in Tfm

males (Vanderschueren, Boonen & Bouillon, 1998). We did not measure energy intake in

Tfm males, and thus it is not known whether this parameter differs from normal WT

males. Examination of activity level found no differences between Tfm and WT males.

Aside from measuring these parameters, one might also treat Tfm males with an

aromatase inhibitor, in order to limit estrogen levels. Finally, gene targeting may also be

employed. Ablation of AR in the brain could result in increased brain estrogen levels.

While a model of AR ablation only in the nervous system was generated very recently

(Juntti et al., 2010), energy balance parameters have not been measured. Thus, we

propose that the unexpected shift in body composition seen in Tfm male rats (increased

Page 130: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

120

LBM% and decreased FBM%) may be due to higher levels of estrogens, which is a

consequence of higher serum T levels. Similar effects are not seen in other rodent models

of androgen-insensitivity (i.e. Tfm mice or ARKO mice), as these strains exhibit

significantly lower levels of circulating T. Such a potentiality represents the major

interplay between androgens, estrogens and body composition.

4.5. Myocyte AR and Oxidative Metabolism

Overall, our results suggest a model whereby increased (or sole) expression of AR

in myocytes is sufficient to induce hypermetabolism, and subsequent improvement of

body composition through reduction of FBM. This is a novel finding, and thus,

elucidation of the mechanism by which myocyte AR is able to alter systemic metabolism

remains an important step.

4.5.1. Local Effects on Skeletal Muscle Metabolism

Our investigation of basal metabolism shows that RMR is heightened in HSA-AR

male rats and mice. This is evidenced by increased O2 consumption in these animals – a

major indicator of elevated systemic metabolism. We also found no differences between

male rats in physical activity, nor was feeding behaviour altered between groups (Rao &

Monks, unpublished data), suggesting that this disparity in RMR was likely the major

driving force behind the changes we see in body composition.

It is likely that these differences might be due to local action of androgens in

skeletal muscle. It is now well understood that alterations in skeletal muscle can strongly

contribute to changes in systemic metabolism (Harrison & Leinwand, 2008). Hypertrophy

Page 131: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

121

of skeletal muscle fibers is associated with increased uptake of glucose and fat, and the

subsequent catabolism of these substrates for the purpose of energy harvest. Therefore, it

is possible that myocyte AR may have local effects on muscle glycogen content. This

possibility was examined in mARKO mice (Ophoff et al., 2009). Here, scientists

examined skeletal muscle glycogen content using biochemical enzymatic fluorometric

assay, and found no significant differences between groups, although a trend toward

lower glycogen content in mARKO males was seen. In addition, treatment of adult rats

with exogenous T has been shown to increase glycogen accumulation in soleus muscle

(Cunha et al., 2005). Furthermore, glycogen content is increased in HSA-AR male mice

(both L78 and L141; Musa et al., In Prep). Here, electron microscopy of skeletal muscle

sections from EDL of HSA-AR animals revealed increased aggregation of glycogen,

while Periodic acid-Schiff (PAS) staining was heightened in HSA-AR EDL sections.

Thus, it appears possible that myocyte AR may have local effects on muscle glycogen

accumulation.

4.5.2. Mitochondrial Biogenesis and Enzyme Activity

Differences in skeletal muscle metabolism are most often linked with muscle

mitochondria, which are the major sources of energy generation in the cell, and are

responsible for catabolism of glucose and fatty acids (Holloszy, 2008). Therefore,

alterations in muscle mitochondrial biogenesis or enzyme activity will likely have major

implications for systemic metabolism and body composition. Recent work from our lab

has shown that muscle mitochondria are significantly altered in HSA-AR male rats and

mice. In HSA-AR male rats, dissected EDL muscles were homogenized and mitochondria

Page 132: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

122

were isolated. Examination of electron transport chain (ETC) enzymes reveals higher

activity of these proteins in HSA-AR males (Fernando et al., 2010). Activity of all four

ETC complexes was heightened in HSA-AR males, and interestingly activity of these

enzymes is also reduced in muscles of Tfm males (as compared to WT males). HSA-

AR/Tfm male rats demonstrate equivalent ETC activity to that seen in WT males,

presumably due to contrasting action of HSA-AR and Tfm on skeletal muscle

mitochondria in these males.

Similar findings are reported in HSA-AR mouse lines (Musa et al., In Prep). ETC

activity is heightened in L78 males, as well as T-treated L78 and L141 females. This

occurs despite significant muscular atrophy in T-treated L141 females. Electron

microscopy of EDL from these animals shows that HSA-AR males (and HSA-AR

females treated with T) demonstrate proliferation of mitochondria. These organelles are

larger in L78 males than WT littermates, and more numerous in L141 males than WT

littermates. Similar results are found in T-treated females of both lines. Although

symptomatic T-treated females and L78 males demonstrate a loss of oxidative fibers,

mitochondrial content appears to be augmented in L78 males. Thus, it seems that

mitochondrial biogenesis and enzyme activity are both increased in HSA-AR mice.

These alterations in myofiber mitochondria provide some insight of a mechanism

by which increased myocyte AR contributes to heightened systemic metabolism and

improved body composition (through increased RMR). Increased myocyte mitochondrial

biogenesis has been associated with increased cellular respiration and β-oxidation of fatty

acids (Cha et al., 2006; Holloszy, 2008). Similarly, increased activity of ETC enzymes in

skeletal muscle has been shown to have similar results (Birch-Machin & Turnbull, 2001).

Page 133: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

123

Reduction in content of skeletal muscle mitochondria is found in patients suffering from

obesity and T2DM (Ritov et al., 2005). Finally, deficits in muscle ETC enzyme activity

are also found in these patient populations (Ritov et al., 2009). Thus, regulation of

skeletal muscle mitochondria has important implications for whole-body physiology and

human health.

The question therefore surrounds exactly how androgens/AR are capable of

modulating mitochondria in skeletal muscle. The most likely potential pathway involves

AR-mediated expression of major factors involved in mitochondrial biogenesis and β-

oxidation (ex: PGC-1α, PPAR family proteins). Unfortunately, very little evidence of

such effects actually exist. Analysis of gene expression in skeletal muscle of ARKO mice

reveals down-regulation of PPARα (Lin et al., 2005). Agonists of this nuclear receptor

have been shown to stimulate mitochondrial fatty acid β-oxidation in skeletal muscle

(Minnich, Tian, Byan & Bilder, 2001). Therefore, it is possible that AR may mediate

expression of skeletal muscle PPARα, thus contributing to normal fatty acid utilization in

myocytes. In addition, it is possible that androgens may be affecting expression of

mitochondrial DNA. Hormone response elements – DNA sequences to which nuclear

receptor/ligand complexes bind in mediating gene expression – have been found in the

mitochondrial genome, suggesting that steroid hormones can act directly upon

mitochondria (Psarra, Solakidi & Sekeris, 2006). More specifically in skeletal muscle,

Weber et al. (2002) showed that treatment of rats with glucocorticoids (another steroid

hormone) resulted in mitochondrial biogenesis. Here these scientists demonstrated that

glucocorticoid receptor was found in skeletal muscle mitochondria, further evidence that

these hormones act directly upon mitochondria. However, to date, similar findings have

Page 134: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

124

not been investigated in the context of androgens, AR and skeletal muscle. In summary,

research surrounding skeletal muscle mitochondria shows that these organelles can be

regulated by muscle AR, although the mechanisms by which this regulation occurs is

largely unknown.

4.5.3. Effects on Other Metabolic Organs

While it is most likely that the differences in body composition and metabolism

between HSA-AR and WT animals are due to direct local actions of androgens in skeletal

muscle, it is important to not forget potential indirect effects on other metabolic organs.

Aside from skeletal muscle, the two most commonly studied tissues known for utilization

of energy substrate (i.e. glucose and fatty acids) are the brain and the liver. Both of these

organs are also known to express AR abundantly (Ruizeveld de Winter et al., 1991).

Whether myocyte AR can have indirect effects on these tissues has not been extensively

studied.

The liver is a major metabolic organ (Kammoun et al., 2009). It regulates glucose

homeostasis through uptake and storage as glycogen, and also helps provide glucose to

major organs that require it (such as the brain and skeletal muscle), through

gluconeogenesis. It is also a major site of fatty acid uptake, lipid storage and β-oxidation.

Recent research reveals that local changes in skeletal muscle can have major implications

for the liver. In a groundbreaking study, Izumiya et al. (2008) report a skeletal muscle

manipulation in transgenic mice that results in a phenotype that is strikingly similar to

that seen due to HSA-AR. Here, scientists use a skeletal muscle-specific, Dox-inducible

transgenic mouse model that overexpresses a constitutively active form of the major

Page 135: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

125

signaling molecule Akt1. Induction of the transgene in adulthood results in selective

hypertrophy of type IIb (fast-twitch, glycolytic) muscle fibers, reduced fat mass, and

increased O2 consumption (and subsequently increased energy expenditure). As

mentioned, these findings largely mimic the effects seen in HSA-AR male rats.

Interestingly, here the scientists found that major oxidative genes (such as PPARγ and

PGC-1α) were down-regulated in skeletal muscle of HSA-AR animals, while major

glycolytic genes were up-regulated. Puzzled by the contrasting increase in systemic

oxidative metabolism, but decreased oxidative gene expression in skeletal muscle, the

authors investigated liver of these animals and found that fatty acid metabolism in this

organ was significantly increased. They hypothesized that this influence on liver

metabolism may occur through systemic action of muscle-secreted factors, termed

‘myokines’. These myokines have been shown to have beneficial effects on body

composition, although their mechanism of action remains unclear (Pederson et al., 2007).

With regards to our results, it is possible that HSA-AR could be operating under a similar

mechanism. Although we did not investigate the liver of HSA-AR males, it may have

played a potential role in β-oxidation of fatty acids, resulting in the reduced adiposity we

see.

The other major target organ for substrate uptake and utilization is the brain.

Whether the brain is involved in mediating the effects of HSA-AR is less likely than the

potential influence of the liver. First, the brain largely does not use fatty acids as an

energy source. Thus, it is highly improbable that it plays a direct role in reducing the

adiposity that we see in HSA-AR. Secondly, relatively little evidence for cross-talk

between skeletal muscle and the brain exists. The little work that has been done in this

Page 136: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

126

field has focused upon efferent influences of the brain on skeletal muscle through the

sympathetic nervous system. For example, the hypothalamus is able to induce

mitochondrial biogenesis in skeletal muscle in response to increased levels of fatty acid

substrate (Cha et al., 2006). That being said, there is some evidence that shows that

skeletal muscle is capable of secreting myokine signals that can act upon the brain and

influence its metabolism (Pederson & Febbraio, 2005). Several muscle-secreted factors

(such as interleukin-6) have been shown to cross the blood-brain barrier (Banks, Castin &

Broadwell, 1995). While interleukin-6 has been shown to have effects on brain

metabolism (Penkowa et al., 2003), these influences have not been well-established, and

thus the influence of muscle on brain metabolism remains hypothetical at best.

In summary, while differences in systemic metabolism and body composition of

HSA-AR male animals may result from local actions of AR in skeletal muscle, it is also

important to remember the roles of other metabolic tissues, and how their influence may

potentially be involved in the effects that we see.

4.6. Comparing HSA-AR Rats and Mice

In order to generalize the role of AR in muscle, we studied two separate lines of

transgenic animals overexpressing AR in this tissue: HSA-AR rats, and HSA-AR L78

mice. Both strains utilize the HSA promoter to drive transgene expression (although a

truncated version is employed in the rats). Comparing the phenotypes of these two species

(elucidated through the work of this study, as well as from previous work) will allow for

greater understanding of what AR does in skeletal muscle.

Page 137: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

127

4.6.1. Phenotype Comparisons

Several similarities between HSA-AR rats and mice exist, but these species are

also marked by major differences. With regards to body composition, similar findings are

seen in both HSA-AR male rats and L78 HSA-AR male mice (as compared to WT

counterparts). Both display increased LBM%, decreased FBM% (including raw FBM)

and reduced BMC. Thus, the existence of these effects in both species suggests that

myocyte AR is capable of improving body composition, primarily by reducing FBM.

Furthermore, study of oxidative metabolism between these two strains also demonstrates

similarities. Both groups show evidence of heightened RMR, indicating that increased

expression of AR in skeletal muscle is capable of generating higher rates of systemic

metabolism. Local examination of skeletal muscle mitochondrial ETC enzymes also

reveals parallel results between strains (Fernando et al., 2010; Musa et al., In Prep). HSA-

AR male rats, L78 male mice, and T-treated L141 female mice all show increased activity

of ETC enzymes in EDL muscle. Thus, taken together, it can be hypothesized that AR

functions in myocytes to aid in regulation of mitochondria, thus resulting in increased

systemic metabolism, and reduction of FBM.

However, there are differences between the two strains that may cloud the details

of this mechanism. Namely, it is demonstrated through HSA-AR mice that

overexpression of AR in muscle can lead to acute muscular pathology and death (Monks

et al., 2007). In contrast, HSA-AR rats show no deleterious effects of transgene

expression. These rats are healthy, and at birth, litters yield the expected 1:1 proportion of

HSA-AR and WT males. Conversely, HSA-AR mice experience robust muscular atrophy,

a phenotype that is more predominant in the L141 line (Monks et al., 2007). The great

Page 138: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

128

majority of males of this line die perinatally. The few that do survive gestation typically

die by PND 10. Although this phenotype is not as severe in L78 male mice,

histopathological analyses of these animals do display some muscle wasting, while we

also found reduced levels of raw LBM in this strain, consistent with mild atrophy.

Ultrastructural analyses reveal that mitochondria in HSA-AR mice are more numerous

and larger, and that myofibril width is narrowed (Musa et al., In Prep). These deleterious

effects in HSA-AR mice complicate the role of AR in muscle, since we find that this

manipulation has largely beneficial effects in our transgenic rats.

4.6.2. Does Expression Level Exaggerate Phenotype?

Despite differences in tissue specificity of expression, the major disparity between

HSA-AR rats and mice exists in the level of AR expression. Western blot analysis

indicates AR expression in EDL muscle of HSA-AR male rats is roughly 2-3 times the

level seen in WT male littermates (Niel et al., 2009). Therefore, the overexpression of AR

in HSA-AR rats is relatively modest. In contrast, the level of overexpression in HSA-AR

mice is significantly higher (Monks et al., 2007). While protein levels were not

quantitatively analyzed in these mice, transgene constructs reveal orders of magnitude

difference in AR copy number, with L78 mice having roughly 100 copies of AR/pg RNA,

and close to 1000 copies of AR/pg RNA in L141 mice. Therefore, the differences we see

between these transgenic strains and species may be related to the level of AR expression

found.

A modest degree of AR overexpression in muscle of rats appears mostly

beneficial, reducing FBM and elevating RMR. A significantly higher level of

Page 139: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

129

overexpression in L78 males seems to induce similar effects; however some

abnormalities in skeletal muscle fibers are now noted, with reduced LBM in these animals

and histopathological indications of muscular atrophy. Finally, boosting overexpression

of AR by another order of magnitude (as is seen in L141 males) results in significant

muscular atrophy, proliferation of mitochondria, aggregation of glycogen and resultant

death.

With these differences in expression level and resultant phenotype in mind,

hypothetical explanations can be put forward in order to account for the compounding

effects of AR in muscle. AR’s most well understood function is as a transcription factor,

regulating the expression of various genes. Previous work in whole-body ARKO mice

reveals that mRNA expression of PPARα (a protein involved in mitochondrial

biogenesis) in skeletal muscle of these animals is significantly reduced (Lin et al., 2005).

Therefore, it is possible that AR in skeletal muscle may be responsible for regulating

expression of proteins involved in mitochondrial biogenesis. Higher and higher amounts

of AR protein would exaggerate this response, thus accounting for the higher levels of

ETC activity and mitochondrial proliferation seen in HSA-AR mice (Musa et al., In

Prep). The subsequent increase in systemic metabolism of these animals would be robust.

When energy expenditure exceeds energy intake, an energy deficit occurs, with

exacerbation of this deficit eventually leading to cell death. This is seen in various models

of neurodegenerative and neuromuscular disease (such as ALS and HD), which are now

being regarded as having significant metabolic components (Chaturvedi et al., 2009). In

fact, scientists are now able to alleviate pathological symptoms in mouse models of these

diseases using creatine supplementation, a muscle-specific energy supplement (Ferrante

Page 140: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

130

et al., 2000; Klivenyi et al., 1999). Thus, while hypermetabolism is often seen as

beneficial, pushing this system to an extreme could easily result in cell death. Similarly,

the modest degree of AR overexpression in HSA-AR rats has largely positive effects in

increasing metabolism and reducing LBM. This effect appears to be exaggerated in HSA-

AR mice, likely due to significantly higher AR expression, and subsequently resulting in

cell death.

4.7. A New Hope: Selective Androgen Receptor Modulators

Our findings may have significant translational potential for application in the

treatment of human disease. Physicians have often prescribed androgen therapy for

patients suffering from sexual dysfunction, as well as other pathologies. However,

previous investigation into androgen therapy shows that the efficacy of these hormones in

a clinical setting is largely limited by their deleterious side effects (Bhasin et al., 2001;

Basaria et al., 2010). These side effects include increased incidence of prostate cancer in

men, masculinization of secondary sexual characteristics in females, and increased

incidence of heart disease in both sexes. This occurs because use of exogenous androgens

causes indiscriminant activation of AR in all tissues. For that reason, a large degree of

investigation in this field has gone into identification of tissue-specific AR that give rise

to the beneficial effects of androgens. By targeting only these AR (and avoiding AR in

prostate and heart), scientists can devise pharmacological agents aimed at treating a

whole host of pathologies, with minimal side effects. Such technology is now on the rise

with the advent of Selective Androgen Receptor Modulators (SARMs), which are a class

of isolated endogenous agents that bind only to AR in specific tissue (Bhasin et al., 2006).

Page 141: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

131

Our research suggests that a SARM targeted toward skeletal muscle might have beneficial

effects in increasing LBM while decreasing FBM, allowing for a powerful treatment

against muscular atrophy and obesity.

4.7.1. Selective Androgen Receptor Modulators

SARMs are not synthetically bioengineered molecules, but are rather endogenous

ligands that are found to bind to AR in specific tissues. For example, 27-

hydroxycholesterol has been found to impair cardiac function not only by forming

plaques in arteries, but also by binding to ER only in cardiovascular tissue and blocking

the beneficial effects of estrogens on vascular function (Umetani et al., 2007). SARMs are

typically not steroid-based, and thus differ from T and steroidal androgens in several

ways. Unlike T, which is converted to active metabolites (namely DHT and estrogen),

nonsteroidal SARMs do not undergo aromatization or 5-α-reduction (Bhasin et al., 2006).

Some specific nonsteroidal SARMs have actually been shown to have more favourable

pharmokinetics, greater AR binding capability, and increased amenability to structural

modifications than their androgen counterparts. SARM technology was first harnessed to

mimic the anabolic functions of androgens in bone, through isolated extraction of

tetrahydroquinoline (Hanada et al., 2003). While treatment of rats with this extract led to

significant improvement in bone density, there were some effects noted in reproductive

tissues. To date, scientists have been able to successfully alter bone composition with

little to no effect on other tissues (Gao & Dalton, 2007).

The potential mechanism of SARM tissue selectivity is still not well understood

(Bhasin et al., 2006). Various mechanisms have been proposed, but none have been

Page 142: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

132

explicitly tested. The most commonly cited mechanism focuses upon alterations in AR

conformation induced by SARMs, and subsequent ability to recruit co-activators (Bhasin

et al., 2006). Binding of T to AR induces conformational change in the ligand-binding

domain of AR. These conformational changes could modulate surface topology and

protein-protein interactions between AR and specific coactivators. Since the androgen

response elements differ between tissues (and the subsequent genes that are regulated also

differ), recruitment of coactivators is specific to particular tissues (Ting & Chang, 2008).

Thus, one can hypothesize that a SARM binds to AR in all tissues, however the

conformational change induced in the AR protein due to SARM binding only facilitates

recruitment of certain tissue-specific coregulators and coactivators. This would lead to

up-regulation (or down-regulation) of androgen responsive genes in specific tissues, and

thus provide a rationale for the tissue-specificity noted through the use of SARMs.

4.7.2. Targeting Muscle

Our research suggests that SARMs created to target myocyte AR may have

significant effects in maintaining (possibly increasing) LBM and decreasing FBM. The

use of SARM technology targeted toward skeletal muscle has already been researched,

for the purposes of harnessing the anabolic effects of androgens on skeletal muscle (Yin

et al., 2003). Gao and colleagues (2005) utilized such a SARM on gonadectomized adult

male rats. They found that treatment of these rats with the SARM increased muscle mass

and bone density. These levels were comparable with those seen in animals treated with

DHT. Interestingly, SARM treatment did not decrease FBM, although DHT did. Similar

findings were reported by Schmidt et al. (2009). Here the authors used a newly isolated

Page 143: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

133

SARM and found that it had anabolic functions on muscle and bone. Microarray analysis

of skeletal muscle showed that treatment with the SARM induced changes in gene

expression that were largely consistent with those induced by DHT. Thus, these findings

suggest that the use of SARMs may have beneficial effects on skeletal muscle and body

composition, but further insight must be gained regarding the mechanism of action by

which these endogenous ligands function.

4.8. Future Directions

Although the findings from our experiments provide us with significant insight

regarding the effects of the HSA-AR transgene and the role of myocyte AR in mediating

whole-body physiology, there are still a tremendous degree of questions that remain

unanswered. Here we will briefly focus on potential avenues for future research, which

would help to further elucidate the function of AR in muscle.

4.8.1. Other Metabolic Parameters in HSA-AR Animals

Our investigation of metabolism in HSA-AR rats and mice largely focused on the

effects of FBM. Once it was demonstrated that HSA-AR animals had significantly

reduced FBM and heightened systemic metabolism, our attention was immediately turned

to local effects of AR on skeletal muscle metabolism. It is now known that AR in skeletal

muscle has significant effects on muscle mitochondria (Fernando et al., 2010; Musa et al,

In Prep.). However, little is known regarding other major metabolic processes in these

animals.

Page 144: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

134

A major contributor to metabolic status is glucose homeostasis. Glucose is the

major fuel of the body, and its catabolism allows for the generation of ATP. Furthermore,

skeletal muscle is known to be one of the major sites of glucose utilization, as it is stored

in this tissue in the form of glycogen, and broken down in muscle mitochondria (Harrison

& Leinwand, 2008). Whole-body ablation of AR in mice results in hyperglycemia and

reduced insulin sensitivity (Lin et al., 2005). With the knowledge that mitochondrial ETC

enzyme activity is significantly enhanced in skeletal muscle of HSA-AR animals, and

since glucose is a major substrate for oxidative phosphorylation, it is reasonable to

assume that glucose parameters may differ between HSA-AR and WT animals. First and

foremost, serum glucose levels can be measured in order to determine if there are any

baseline differences between groups. Animals can also be tested on a glucose tolerance

test, where glucose is administered (either orally or via injection), and blood glucose rates

are then measured. Faster rates of glucose clearance (marked by larger decreases in blood

glucose after glucose administration) suggest increased insulin sensitivity and higher

glucose uptake by target tissues (Chen et al., 2003). Furthermore, expression of major

glucose regulatory proteins (such as GLUT4) could be measured. It is possible that AR in

muscle might be involved in increasing expression of GLUT4 (or FATP) and thus

increasing clearance of glucose and fatty acids from the blood. Such a finding would help

in explaining the higher aggregation of glycogen found in HSA-AR L78 and L141 mice

(Musa et al., In Prep). It is now understood that T2DM and obesity are inextricably linked

(Zitzmann, 2009), and thus treatment of one disease is typically helpful in treating the

other.

Page 145: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

135

Examination of liver in HSA-AR animals will also be important. Accumulation of

fatty acids in the liver (hepatic steatosis) is a major cause of liver disease, and its

incidence is significantly increased in individuals with higher levels of FBM (Kammoun

et al., 2009). Furthermore, Izumiya et al. (2008) reported a manipulation in skeletal

muscle of HSA-AR mice that did not have local effects on skeletal muscle oxidative

capacity, but rather increased β-oxidation by the liver. The findings reported in that study

(selective hypertrophy of fast-twitch, glycolytic muscle fibers with concomitant decrease

in fat mass and improved metabolic parameters) strongly parallel those seen in HSA-AR

rats (Fernando et al., 2010). While we do find local effects in metabolic function of

skeletal muscle, investigation of gene expression and oxidative capacity in liver could

provide an alternative route by which fatty acid β-oxidation may be occurring.

Furthermore, histological analysis of fatty acids in hepatocytes may indicate whether

myocyte AR can reduce the incidence (or magnitude) of hepatic steatosis when animals

are placed on a high-fat diet. A similar effect has already been shown in mice, where

knockout of AR only in hepatocytes results in hepatic steatosis (Lin et al., 2008). This

finding highlights the role of AR in fatty acid metabolism.

4.8.2. Identifying AR and Mitochondrial Interactions in Skeletal Muscle

As noted, work from our lab has demonstrated that expression of HSA-AR in

skeletal muscle has significant local effects on muscle mitochondria. This includes

increase in ETC enzyme activity in HSA-AR rats, coupled with reduced activity in Tfm

rats (Fernando et al., 2010) and similar effects on enzyme activity with increased

mitochondrial proliferation in HSA-AR mice (Musa et al., In Prep). These findings are

Page 146: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

136

interesting, particularly because it has been shown that there are sex differences in

expression of mitochondrial proteins in skeletal muscle of adult humans, with higher

expression levels found in men (Welle, Tawil & Thornton, 2008). Whether these

differences are entirely due to androgens is unclear. Nevertheless, identification of the

mechanisms by which AR is able modulate mitochondria represent an important step in

elucidation of AR’s role in skeletal muscle. However, to date it is not know whether AR

even binds and colocalizes to mitochondria in skeletal muscle myocytes. Solving this

unknown will provide information as to whether AR acts directly upon muscle

mitochondria.

Previous studies have also linked AR to mitochondrial function, with

demonstration of AR-mitochondria colocalization in non-muscle tissues. Ranganathan

and colleagues (2009) found that AR possessing a polyglutamine expansion mutation (as

seen in models of SBMA) results in altered expression of various mitochondrial proteins,

as well as mitochondrial permeability. This study also found that AR associates with

mitochondria in cultured PG12 cells. Transmission electron microscopy showed that AR

localized within mitochondria as well as along the membrane. However, it is unclear as to

whether this modulatory effect is due to indirect effects on the transcription of

mitochondrial genes encoded in the nucleus, or direct effects of the mutant AR protein on

mitochondria (or both). Further to this, AR has been detected in mitochondria of LNCaP

cells (androgen-sensitive human prostate adenocarcinoma cells) as well as the midpiece

region of human sperm cells (Solakidi et al., 2005). It remains unclear as to what function

AR may perform in the mitochondria of these cells. It has been hypothesized that these

receptors may act by upregulating the transcription of various mitochondrial genes, as

Page 147: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

137

well as genes involved in oxidative phosphorylation (Psarra & Sekeris, 2008). This has

been demonstrated in skeletal muscle, where other nuclear receptors (namely

glucocorticoid receptor) have been shown to be involved in mitochondrial biogenesis

(Weber et al., 2002). Glucocorticoid receptor expression has also been found in

mitochondria (Psarra et al., 2006).

There are several methods that can be used to determine if AR colocalizes with

skeletal muscle mitochondria. This can be done mostly simply through isolation of

skeletal muscle mitochondria and then subsequently probing for AR in the mitochondria

using an immunoblot. A positive signal would suggest the existence of an AR-

mitochondria complex. This could alternatively be accomplished through immunogold

labeling for AR, followed by electron microscopy, as employed by Ranganathan et al.

(2009). The benefits of this method are the increased level of resolution obtained, and the

fact that quantification can be achieved through counting of AR-positive mitochondria.

Determining whether AR colocalizes with mitochondria is an important step in

elucidating the mechanisms by which myocyte AR can influence this organelle.

4.8.3. Molecular and Biochemical Assays of Key Metabolic Players

AR’s role as a transcription factor suggests that this nuclear receptor might

influence body composition by altering expression of various metabolic genes. Thus,

gene expression assays will be especially useful for potentially determining what muscle-

specific factors are involved in regulating the effects that we see on body composition of

HSA-AR animals. The most well understood regulator of skeletal muscle metabolism is

the PPARγ coactivator PGC-1α, with increased expression of skeletal muscle PGC-1α

Page 148: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

138

being associated with mitochondrial biogenesis, increased systemic metabolism, and

reduction of FBM (Calvo et al., 2008; Wenz et al., 2009). Similar effects are known to

occur for PPARα, a gene whose expression is reduced in skeletal muscle of ARKO mice

(Lin et al., 2005). Thus, examination of expression of these and other important metabolic

genes in skeletal muscle of HSA-AR animals could provide significant insight into the

mechanisms by which mitochondrial biogenesis and reduced FBM occur. Analysis of

gene expression could be accomplished using traditional methods of examining mRNA,

such as Quantitative PCR and Northern blot analyses, along with quantification of protein

expression through Western blot.

Alternatively, it is possible that AR may exert its action through nongenomic

actions. In a nongenomic mechanism, binding of AR by its ligand results in formation of

secondary messengers and influence of the secondary messengers on cell signaling. For

example, glial cell culture experiments show that activation of AR by DHT results in

increased phosphorylation of both ERK and Akt, key effectors in the MAPK and PI3-

kinase signaling pathways, and that phosphorylation of these molecules in inhibited by

administration of an AR antagonist (Gatson, Kaur & Singh, 2006). Such effects can be

contrasted with the classical role of AR in regulation of gene expression. Several lines of

evidence indicate that AR functions using such nongenomic mechanisms in adipose tissue

(Mayes & Watson, 2004). Exhibition of such mechanisms in skeletal muscle by AR have

not been found. However, it is nevertheless possible that AR may exert such effects on

key metabolic signaling molecules in skeletal muscle, such as AMPK. Activation of

AMPK and phosphorylation of its targets has shown to be an important process capable

of regulating mitochondrial biogenesis and fiber type switching in skeletal muscle, as

Page 149: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

139

well as increasing metabolic parameters (de Lange et al., 2007; Narkar et al., 2008). We

can evaluate activation of AMPK through the use of biochemical assays, and detection of

phosphorylation of AMPK targets. Administration of AR antagonists will indicate

whether any such effects are T-dependent.

4.9. Conclusions

This study revealed the changes in body composition and metabolism associated

with overexpression of AR in muscle fibers. More specifically, we find that

overexpression of AR in muscle increased LBM%, and decreased FBM, FBM% and

BMC. These findings are seen in both rat and mouse HSA-AR strains. Furthermore, all of

these findings are seen in male rats only expressing AR in muscle fibers, indicating that

androgen activity in this cellular population is sufficient for induction of the effects that

we see. Treatment of female rats with exogenous T recapitulates our findings in males,

suggesting that they are likely dependent upon the androgen ligand, and are not simply

the result of transgene expression only.

Perhaps most interesting, we show that AR in myocytes is sufficient for

androgenic reduction of FBM, which is confirmed through dissection of individual fat

pads, and analysis of adipocyte size. This is a novel finding, as androgens are typically

thought to reduce FBM by acting upon AR in adipocytes, and inhibiting adipogenesis

during development (Singh et al., 2003; Singh et al., 2006). However, our findings

explain why ablation of AR only in adipocytes (Yu et al., 2008) is incapable of

recapitulating the adult-onset obesity found in whole body ARKO (Lin et al., 2005; Fan et

al., 2005). Investigation of metabolic parameters shows that our HSA-AR males exhibit

Page 150: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

140

heightened resting metabolism, which is complimentary to the reduced RMR values

found in ARKO mice (Fan et al., 2005). Future research should focus upon elucidation of

the mechanisms involved in heightened metabolism. Recent work from our lab shows that

AR is capable of modulating skeletal muscle mitochondria (Fernando et al., 2010; Musa

et al., In Prep). Therefore, a more comprehensive analysis of how AR is capable of

regulating mitochondria in muscle will provide greater insight into how androgens and

AR influence body composition and energy homeostasis.

Page 151: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

141

References

Abel, A., Walcott, J., Woods, J., Duda, J., & Merry, D. E. (2001). Expression of expanded repeat androgen receptor produces neurologic disease in transgenic mice. Human Molecular Genetics, 10(2), 107-116.

Adams, G. R., Hather, B. M., Baldwin, K. M., & Dudley, G. A. (1993). Skeletal muscle myosin heavy chain composition and resistance training. Journal of Applied Physiology, 74(2), 911-115 Allen, D. G., Lamb, G. D., & Westerblad, H. (2008). Skeletal muscle fatigue: cellular mechanisms. Physiological Reviews, 88(1), 287-332. Aizawa, K., Iemitsu, M., Otsuki, T., Maeda, S., Miyauchi, T., & Mesaki, N. (2008). Sex

differences in steroidogenesis in skeletal muscle following a single bout of exercise in rats. Journal of Applied Physiology, 104(1), 67-74.

Altuwaijri, S., Chuang, K., Lai, K., Lai, J., Lin, H., Young, F. M., et al. (2009). Susceptibility to autoimmunity and B cell resistance to apoptosis in mice lacking androgen receptor in B cells. Molecular Endocrinology, 23(4), 444-453.

Araki, E., Lipes, M. A., Patti, M. E., Brüning, J. C., Haag, B., Johnson, R. S., & Kahn, C. R. (1994). Alternative pathway of insulin signalling in mice with targeted disruption of the IRS-1 gene. Nature, 372(6502), 186-190.

Arita, Y., Kihara, S., Ouchi, N., Takahashi, M., Maeda, K., Miyagawa, J., Hotta, K., et al. (1999). Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochemical and Biophysical Research Communications, 257(1), 79-83.

Asarian, L., & Geary, N. (2002). Cyclic estradiol treatment normalizes body weight and restores physiological patterns of spontaneous feeding and sexual receptivity in ovariectomized rats. Hormones and Behavior, 42(4), 461-471.

Asarian, L., & Geary, N. (2006). Modulation of appetite by gonadal steroid hormones. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 361(1471), 1251-1263.

Atsuta, N., Watanabe, H., Ito, M., Banno, H., Suzuki, K., Katsuno, M., Tanaka, F., et al. (2006). Natural history of spinal and bulbar muscular atrophy (SBMA): a study of 223 Japanese patients. Brain, 129(Pt 6), 1446-1455.

Bailey, C. K., Andriola, I. F. M., Kampinga, H. H., & Merry, D. E. (2002). Molecular chaperones enhance the degradation of expanded polyglutamine repeat androgen receptor in a cellular model of spinal and bulbar muscular atrophy. Human Molecular Genetics, 11(5), 515-523.

Banks, W. A., Kastin, A. J., & Broadwell, R. D. (1995). Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation, 2(4), 241-248.

Barnes, B. R., Long, Y. C., Steiler, T. L., Leng, Y., Galuska, D., Wojtaszewski, J. F. P., Andersson, L., et al. (2005). Changes in exercise-induced gene expression in 5'- AMP-activated protein kinase gamma3-null and gamma3 R225Q transgenic mice. Diabetes, 54(12), 3484-3489.

Baron, S., Manin, M., Beaudoin, C., Leotoing, L., Communal, Y., Veyssiere, G., & Morel, L. (2004). Androgen receptor mediates non-genomic activation of phosphatidylinositol 3-OH kinase in androgen-sensitive epithelial cells. The Journal of Biological Chemistry, 279, 14579-14586.

Page 152: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

142

Barrionuevo, F., Bagheri-Fam, S., Klattig, J., Kist, R., Taketo, M. M., Englert, C., & Scherer, G. (2006). Homozygous inactivation of Sox9 causes complete XY sex reversal in mice. Biology of Reproduction, 74(1), 195-201.

Barske, L. A., & Capel, B. (2008). Blurring the edges in vertebrate sex determination. Current Opinion in Genetics & Development, 18(6), 499-505.

Basaria, S., Coviello, A. D., Travison, T. G., Storer, T. W., Farwell, W. R., Jette, A. M., Eder, R., et al. (2010). Adverse events associated with testosterone administration. The New England Journal of Medicine, 363(2), 109-122.

Bhasin, S., Calof, O. M., Storer, T. W., Lee, M. L., Mazer, N. A., Jasuja, R., et al. (2006). Drug insight: Testosterone and selective androgen receptor modulators as anabolic therapies for chronic illness and aging. Nature Clinical Practice. Endocrinology & Metabolism, 2(3), 146-159.

Bhasin, S., Storer, T. W., Berman, N., Callegari, C., Clevenger, B., Phillips, J., et al. (1996). The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. The New England Journal of Medicine, 335(1), 1-7.

Bhasin, S., Storer, T. W., Berman, N., Yarasheski, K. E., Clevenger, B., Phillips, J., Lee, W. P., et al. (1997). Testosterone replacement increases fat-free mass and muscle size in hypogonadal men. The Journal of Clinical Endocrinology and Metabolism, 82(2), 407-413.

Bhasin, S., Woodhouse, L., Casaburi, R., Singh, A. B., Bhasin, D., Berman, N., Chen, X., et al. (2001). Testosterone dose-response relationships in healthy young men. American Journal of Physiology. Endocrinology and Metabolism, 281(6), E1172- 1181.

Biddinger, S. B., & Kahn, C. R. (2006). From mice to men: insights into the insulin resistance syndromes. Annual Review of Physiology, 68, 123-158.

Bilezikian, J. P. (2002). Sex steroids, mice, and men: when androgens and estrogens get very close to each other. Journal of Bone and Mineral Research, 17(4), 563-566.

Billington, C. J., Briggs, J. E., Harker, S., Grace, M., & Levine, A. S. (1994). Neuropeptide Y in hypothalamic paraventricular nucleus: a center coordinating energy metabolism. The American Journal of Physiology, 266(6 Pt 2), R1765- 1770.

Birch-Machin, M. A., & Turnbull, D. M. (2001). Assaying mitochondrial respiratory complex activity in mitochondria isolated from human cells and tissues. Methods in Cell Biology, 65, 97-117.

Björntorp, P. (1997). Obesity. Lancet, 350(9075), 423-426. Björntorp, P. (1997). Body fat distribution, insulin resistance, and metabolic diseases.

Nutrition, 13(9), 795-803. Bland, R. (2000). Steroid hormone receptor expression and action in bone. Clinical

Science, 98(2), 217-240. Bojesen, A., Kristensen, K., Birkebaek, N. H., Fedder, J., Mosekilde, L., Bennett, P.,

Laurberg, P., et al. (2006). The metabolic syndrome is frequent in Klinefelter's syndrome and is associated with abdominal obesity and hypogonadism. Diabetes Care, 29(7), 1591-1598.

Breedlove, S. M., & Arnold, A. P. (1980). Hormone accumulation in a sexually dimorphic motor nucleus of the rat spinal cord. Science, 210(4469), 564-566. Breedlove, S. M., & Arnold, A. P. (1981). Sexually dimorphic motor nucleus in the rat

Page 153: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

143

spinal cord: response to adult hormone manipulation, absence in androgen- insensitive rats. Brain Research, 225(2), 297-307.

Breedlove, S. M., & Arnold, A. P. (1983). Hormonal control of a developing neuromuscular system. I. Complete demasculinization of the male rat spinal nucleus of the bulbocavernosus using the anti-androgen flutamide. The Journal of Neuroscience, 3(2), 417-423.

Breedlove, S. M., Jacobson, C. D., Gorski, R. A., & Arnold, A. P. (1982). Masculinization of the female rat spinal cord following a single neonatal injection of testosterone propionate but not estradiol benzoate. Brain Research, 237(1), 173-181. Brennan, K. J., & Hardeman, E. C. (1993). Quantitative analysis of the human alpha-

skeletal actin gene in transgenic mice. The Journal of Biological Chemistry, 268(1), 719-725.

Bonen, A., Parolin, M. L., Steinberg, G. R., Calles-Escandon, J., Tandon, N. N., Glatz, J. F., et al. (2004). Triacyglycerol accumulation in human obesity and type 2 diabetes is associated with increased rates of skeletal muscle fatty acid transport and increased sarcolemmal FAT/CD36. The FASEB Journal, 18(10), 1144-1146. Bouzakri, K., Zachrisson, A., Al-Khalili, L., Zhang, B. B., Koistinen, H. A., Krook, A., &

Zierath, J. R. (2006). siRNA-based gene silencing reveals specialized roles of IRS-1/Akt2 and IRS-2/Akt1 in glucose and lipid metabolism in human skeletal muscle. Cell Metabolism, 4(1), 89-96.

Cadigan, K. M., & Nusse, R. (1997). Wnt signaling: a common theme in animal development. Genes & Development, 11(24), 3286-3305.

Callewaert, F., Venken, K., Ophoff, J., De Gendt, K., Torcasio, A., van Lenthe, G. H., et al. (2009). Differential regulation of bone and body composition in male mice with combined inactivation of androgen and estrogen receptor-alpha. The FASEB Journal, 23(1), 232-240.

Calvo, J. A., Daniels, T. G., Wang, X., Paul, A., Lin, J., Spiegelman, B. M., et al. (2008). Muscle-specific expression of PPARgamma coactivator-1alpha improves exercise performance and increases peak oxygen uptake. Journal of Applied Physiology, 104(5), 1304-1312.

Caplice, N. M., & Deb, A. (2004). Myocardial-cell replacement: the science, the clinic and the future. Nature Clinical Practice. Cardiovascular Medicine, 1(2), 90-95. Cha, S., Rodgers, J. T., Puigserver, P., Chohnan, S., & Lane, M. D. (2006). Hypothalamic

malonyl-CoA triggers mitochondrial biogenesis and oxidative gene expression in skeletal muscle: Role of PGC-1alpha. Proceedings of the National Academy of Sciences of the United States of America, 103(42), 15410-15415.

Chambon, C., Duteil, D., Vignaud, A., Ferry, A., Messaddeq, N., Malvindi, R., et al. (2010). Myocytic androgen receptor controls the strength but not the mass of limb muscles. Proceedings of the National Academy of Sciences of the United States of America, 107(32), 14327-14332.

Chang, C. S., Kokontis, J., & Liao, S. T. (1988). Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science, 240(4850), 324-326.

Chang, C., Saltzman, A., Yeh, S., Young, W., Keller, E., Lee, H. J., Wang, C., et al. (1995). Androgen receptor: an overview. Critical Reviews in Eukaryotic Gene Expression, 5(2), 97-125.

Page 154: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

144

Chaturvedi, R. K., Adhihetty, P., Shukla, S., Hennessy, T., Calingasan, N., Yang, L., et al. (2009). Impaired PGC-1alpha function in muscle in Huntington's disease. Human Molecular Genetics, 18(16), 3048-3065.

Chawla, A., Repa, J. J., Evans, R. M., & Mangelsdorf, D. J. (2001). Nuclear receptors and lipid physiology: opening the X-files. Science, 294(5548), 1866-1870.

Chen, Z. P., Stephens, T. J., Murthy, S., Canny, B. J., Hargreaves, M., Witters, L. A., et al. (2003). Effect of exercise intensity on skeletal muscle AMPK signaling in humans. Diabetes, 52(9), 2205-2212. Chuang, K., Altuwaijri, S., Li, G., Lai, J., Chu, C., Lai, K., et al. (2009). Neutropenia

with impaired host defense against microbial infection in mice lacking androgen receptor. The Journal of Experimental Medicine, 206(5), 1181-1199.

Claessens, F., Verrijdt, G., Schoenmakers, E., Haelens, A., Peeters, B., Verhoeven, G., & Rombauts, W. (2001). Selective DNA binding by the androgen receptor as a mechanism for hormone-specific gene regulation. The Journal of Steroid Biochemistry and Molecular Biology, 76(1-5), 23-30.

Clapham, J. C., & Arch, J. R. S. (2007). Thermogenic and metabolic antiobesity drugs: rationale and opportunities. Diabetes, Obesity & Metabolism, 9(3), 259-275.

Clapham, J. C., Arch, J. R., Chapman, H., Haynes, A., Lister, C., Moore, G. B., et al. (2000). Mice overexpressing human uncoupling protein-3 in skeletal muscle are hyperphagic and lean. Nature, 406(6794), 415-418.

Clegg, D. J., Brown, L. M., Woods, S. C., & Benoit, S. C. (2006). Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes, 55(4), 978-987.

Coen, P. M., Dube, J. J., Amati, F., Stefanovic-Racic, M., Ferrell, R. E., Toledo, F. G., et al. (2010). Insulin resistance is associated with higher intramyocellular triglycerides in type I but not type II myocytes concomitant with higher ceramide content. Diabetes, 59(1), 80-88. Compston, J. E. (2001). Sex steroids and bone. Physiological Reviews, 81(1), 419-447. Constable, S. H., Favier, R. J., McLane, J. A., Fell, R. D., Chen, M., & Holloszy, J. O. (1987). Energy metabolism in contracting rat skeletal muscle: adaption to exercise training. American Journal of Physiology, 253(2 Pt 1), C316-C322. Cooke, B. M., Tabibnia, G., & Breedlove, S. M. (1999). A brain sexual dimorphism

controlled by adult circulating androgens. Proceedings of the National Academy of Sciences of the United States of America, 96(13), 7538-7540.

Corva, P. M., & Medrano, J. F. (2000). Diet effects on weight gain and body composition in high growth (hg/hg) mice. Physiological Genomics, 3(1), 17-23.

Couse, J. F., & Korach, K. S. (1999). Estrogen receptor null mice: what have we learned and where will they lead us? Endocrine Reviews, 20(3), 358-417.

Cunha, T. S., Tanno, A. P., Costa Sampaio Moura, M. J., & Marcondes, F. K. (2005). Influence of high-intensity exercise training and anabolic androgenic steroid treatment on rat tissue glycogen content. Life Sciences, 77(9), 1030-1043.

Coggan, A. R., Kohrt, W. M., Spina, R. J., Bier, D. M., & Holloszy, J. O. (1990). Endurance training decreases plasma glucose turnover and oxidation during moderate-intensity exercise in men. The Journal of Applied Physiology, 68(3), 990-996.

Page 155: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

145

de Lange, P., Moreno, M., Silvestri, E., Lombardi, A., Goglia, F., & Lanni, A. (2007). Fuel economy in food-deprived skeletal muscle: signaling pathways and regulatory mechanisms. The FASEB Journal, 21(13), 3431-3441.

Derby, C. A., Zilber, S., Brambilla, D., Morales, K. H., & McKinlay, J. B. (2006). Body mass index, waist circumference and waist to hip ratio and change in sex steroid hormones: the Massachusetts Male Ageing Study. Clinical Endocrinology, 65(1), 125-131.

Dieudonne, M. N., Pecquery, R., Boumediene, A., Leneveu, M. C., & Giudicelli, Y. (1998). Androgen receptors in human preadipocytes and adipocytes: regional specificities and regulation by sex steroids. The American Journal of Physiology, 274(6 Pt 1), C1645-1652.

Dieudonne, M. N., Pecquery, R., Leneveu, M. C., & Giudicelli, Y. (2000). Opposite effects of androgens and estrogens on adipogenesis in rat preadipocytes: evidence for sex and site-related specificities and possible involvement of insulin-like growth factor 1 receptor and peroxisome proliferator-activated receptor gamma2. Endocrinology, 141(2), 649-656.

DiNapoli, L., & Capel, B. (2008). SRY and the standoff in sex determination. Molecular Endocrinology, 22(1), 1-9.

Döhler, K. D., Coquelin, A., Davis, F., Hines, M., Shryne, J. E., & Gorski, R. A. (1984). Pre- and postnatal influence of testosterone propionate and diethylstilbestrol on differentiation of the sexually dimorphic nucleus of the preoptic area in male and female rats. Brain Research, 302(2), 291-295.

Doumit, M. E., Cook, D. R., & Merkel, R. A. (1996). Testosterone up-regulates androgen receptors and decreases differentiation of porcine myogenic satellite cells in vitro. Endocrinology, 137(4), 1385-1394. Dudley, G. A., Tullson, P. C., & Terjung, R. L. (1987). Influence of mitochondrial content on the sensitivity of respiratory control. The Journal of Biological Chemistry, 262(19), 9109-9114. Dunaif, A., Graf, M., Mandeli, J., Laumas, V., & Dobrjansky, A. (1987). Characterization

of groups of hyperandrogenic women with acanthosis nigricans, impaired glucose tolerance, and/or hyperinsulinemia. The Journal of Clinical Endocrinology and Metabolism, 65(3), 499-507.

Dupuis, L., Oudart, H., René, F., Gonzalez de Aguilar, J., & Loeffler, J. (2004). Evidence for defective energy homeostasis in amyotrophic lateral sclerosis: benefit of a high-energy diet in a transgenic mouse model. Proceedings of the National Academy of Sciences of the United States of America, 101(30), 11159-11164.

el-Migdadi, F., Alfano, J., Brownie, A. C., & Gallant, S. (1996). Cytochrome c oxidase in rat adrenal and liver: effects of anti-androgen treatment and studies in testicular feminized rats. Endocrine Research, 22(1), 59-75.

Elias, C. F., Lee, C., Kelly, J., Aschkenasi, C., Ahima, R. S., Couceyro, P. R., Kuhar, M. J., et al. (1998). Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron, 21(6), 1375-1385.

Elmquist, J. K., Elias, C. F., & Saper, C. B. (1999). From lesions to leptin: hypothalamic control of food intake and body weight. Neuron, 22(2), 221-232.

Emmelot-Vonk, M. H., Verhaar, H. J. J., Nakhai Pour, H. R., Aleman, A., Lock, T. M. T. W., Bosch, J. L. H. R., et al. (2008). Effect of testosterone supplementation on

Page 156: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

146

functional mobility, cognition, and other parameters in older men: a randomized controlled trial. JAMA: The Journal of the American Medical Association, 299(1), 39-52.

Enerbäck, S. (2010). Human brown adipose tissue. Cell Metabolism, 11(4), 248-252. Enerbäck, S., Jacobsson, A., Simpson, E. M., Guerra, C., Yamashita, H., Harper, M. E., &

Kozak, L. P. (1997). Mice lacking mitochondrial uncoupling protein are cold- sensitive but not obese. Nature, 387(6628), 90-94.

Eriksson, A., Kadi, F., Malm, C., & Thornell, L. E. (2005). Skeletal muscle morphology of power-lifters with and without anabolic steroids. Histochemistry and Cell Biology, 124(2), 167-175. Fairchild, T. J., & Fournier, P. A. (2004). Glycogen determination using periodic acid-

schiff: artifact of muscle preparation. Medicine and Science in Sports and Exercise, 36(12), 2053-2058.

Fan, W., Boston, B. A., Kesterson, R. A., Hruby, V. J., & Cone, R. D. (1997). Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature, 385(6612), 165-168.

Fan, W., Yanase, T., Nomura, M., Okabe, T., Goto, K., Sato, T., et al. (2005). Androgen receptor null male mice develop late-onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion. Diabetes, 54(4), 1000-1008.

Fernando, S. M., Rao, P., Niel, L., Chatterjee, D., Stagljar, M., & Monks, D. A. (2010). Myocyte androgen receptors increase metabolic rate and improve body composition by reducing fat mass. Endocrinology, 151(7), 3125-3132. Ferrante, R. J., Andreassen, O. A., Jenkins, B. G., Dedeoglu, A., Kuemmerle, S., Kubilus,

J. K., Kaddurah-Daouk, R., et al. (2000). Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease. The Journal of Neuroscience, 20(12), 4389-4397.

Fischbeck, K. H. (1997). Kennedy disease. Journal of Inherited Metabolic Disease, 20(2), 152-158.

Fischbeck, K. H., Ionasescu, V., Ritter, A. W., Ionasescu, R., Davies, K., Ball, S., Bosch, P., et al. (1986). Localization of the gene for X-linked spinal muscular atrophy. Neurology, 36(12), 1595-1598.

Fisher, C. R., Graves, K. H., Parlow, A. F., & Simpson, E. R. (1998). Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proceedings of the National Academy of Sciences of the United States of America, 95(12), 6965-6970.

Fishman, R. B., & Breedlove, S. M. (1992). Local perineal implants of anti-androgen block masculinization of the spinal nucleus of the bulbocavernosus. Brain Research. Developmental Brain Research, 70(2), 283-286. Fix, C., Jordan, C., Cano, P., & Walker, W. H. (2004). Testosterone activates mitogen-

activated protein kinase and the cAMP response element binding protein transcription factor in Sertoli cells. Proceedings of the National Academy of Sciences of the United States of America, 101(30), 10919-10924.

Floyd, N. F., Medes, G., & Weinhouse, S. (1947). Fatty acid metabolism; conversion of acetoacetate to citrate in animal tissues, studied with isotopic carbon. The Journal of Biological Chemistry, 171(2), 633-638.

Page 157: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

147

Ford, E. S., Giles, W. H., & Dietz, W. H. (2002). Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA: The Journal of the American Medical Association, 287(3), 356-359.

Friedman, J. M. (2009). Obesity: Causes and control of excess body fat. Nature, 459(7245), 340-342.

Friedman, J. M., & Halaas, J. L. (1998). Leptin and the regulation of body weight in mammals. Nature, 395(6704), 763-770.

Gambacciani, M., Ciaponi, M., Cappagli, B., Piaggesi, L., De Simone, L., Orlandi, R., & Genazzani, A. R. (1997). Body weight, body fat distribution, and hormonal replacement therapy in early postmenopausal women. The Journal of Clinical Endocrinology and Metabolism, 82(2), 414-417.

Garcia, E., Lacasa, M., Agli, B., Giudicelli, Y., & Lacasa, D. (1999). Modulation of rat preadipocyte adipose conversion by androgenic status: involvement of C/EBPs transcription factors. The Journal of Endocrinology, 161(1), 89-97.

Garcia-Roves, P. M., Osler, M. E., Holmström, M. H., & Zierath, J. R. (2008). Gain-of- function R225Q mutation in AMP-activated protein kinase gamma3 subunit increases mitochondrial biogenesis in glycolytic skeletal muscle. The Journal of Biological Chemistry, 283(51), 35724-35734.

Gao, Q., Mezei, G., Nie, Y., Rao, Y., Choi, C. S., Bechmann, I., Leranth, C., et al. (2007). Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nature Medicine, 13(1), 89-94.

Gao, W., Reiser, P. J., Coss, C. C., Phelps, M. A., Kearbey, J. D., Miller, D. D., et al. (2005). Selective androgen receptor modulator treatment improves muscle strength and body composition and prevents bone loss in orchidectomized rats. Endocrinology, 146(11), 4887-4897.

Gatson, J. W., Kaur, P., & Singh, M. (2006). Dihydrotestosterone differentially modulates the mitogen-activated protein kinase and the phosphoinositide 3- kinase/Akt pathways through the nuclear and novel membrane androgen receptor in C6 cells. Endocrinology, 147(4), 2028-2034.

Gehlert, D. R. (1999). Role of hypothalamic neuropeptide Y in feeding and obesity. Neuropeptides, 33(5), 329-338.

Gloaguen, I., Costa, P., Demartis, A., Lazzaro, D., Di Marco, A., Graziani, R., Paonessa, G., et al. (1997). Ciliary neurotrophic factor corrects obesity and diabetes associated with leptin deficiency and resistance. Proceedings of the National Academy of Sciences of the United States of America, 94(12), 6456-6461.

Gori, F., Hofbauer, L. C., Conover, C. A., & Khosla, S. (1999). Effects of androgens on the insulin-like growth factor system in an androgen-responsive human osteoblastic cell line. Endocrinology, 140(12), 5579-5586.

Guigon, C. J., & Magre, S. (2006). Contribution of germ cells to the differentiation and maturation of the ovary: insights from models of germ cell depletion. Biology of Reproduction, 74(3), 450-458.

Gupta, V., Bhasin, S., Guo, W., Singh, R., Miki, R., Chauhan, P., et al. (2008). Effects of dihydrotestosterone on differentiation and proliferation of human mesenchymal stem cells and preadipocytes. Molecular and Cellular Endocrinology, 296(1-2), 32-40.

Page 158: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

148

Haarbo, J., Marslew, U., Gotfredsen, A., & Christiansen, C. (1991). Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause. Metabolism, 40(12), 1323-1326.

Halaas, J. L., Gajiwala, K. S., Maffei, M., Cohen, S. L., Chait, B. T., Rabinowitz, D., Lallone, R. L., et al. (1995). Weight-reducing effects of the plasma protein encoded by the obese gene. Science, 269(5223), 543-546.

Hanada, K., Furuya, K., Yamamoto, N., Nejishima, H., Ichikawa, K., Nakamura, T., Miyakawa, M., et al. (2003). Bone anabolic effects of S-40503, a novel nonsteroidal selective androgen receptor modulator (SARM), in rat models of osteoporosis. Biological & Pharmaceutical Bulletin, 26(11), 1563-1569.

Hancock, C. R., Han, D., Chen, M., Terada, S., Yasuda, T., Wright, D. C., et al. (2008). High-fat diets cause insulin resistance despite an increase in muscle mitochondria. Proceedings of the National Academy of Sciences of the United States of America, 105(22), 7815-7820.

Harrison, B. C., & Leinwand, L. A. (2008). Fighting fat with muscle: bulking up to slim down. Cell Metabolism, 7(2), 97-98.

Hartgens, F., & Kuipers, H. (2004). Effects of androgenic-anabolic steroids in athletes. Sports Medicine, 34(8), 513-554. Hattner, R., Epker, B. N., & Frost, H. M. (1965). Suggested sequential mode of control of

changes in cell behaviour in adult bone remodelling. Nature, 206(983), 489-490. He, B., Bai, S., Hnat, A. T., Kalman, R. I., Minges, J. T., Patterson, C., & Wilson, E. M.

(2004). An androgen receptor NH2-terminal conserved motif interacts with the COOH terminus of the Hsp70-interacting protein (CHIP). The Journal of Biological Chemistry, 279(29), 30643-30653.

Heine, P. A., Taylor, J. A., Iwamoto, G. A., Lubahn, D. B., & Cooke, P. S. (2000). Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proceedings of the National Academy of Sciences of the United States of America, 97(23), 12729-12734.

Heinlein, C. A., & Chang, C. (2002). The roles of androgen receptors and androgen- binding proteins in nongenomic androgen actions. Molecular Endocrinology, 16(10), 2181-2187.

Hegarty, B. D., Cooney, G. J., Kraegen, E. W., & Furler, S. M. (2002). Increased efficiency of fatty acid uptake contributes to lipid accumulation in skeletal muscle of high-fat fed insulin-resistant rats. Diabetes, 51(5), 1477-1484. Hofbauer, L. C., & Khosla, S. (1999). Androgen effects on bone metabolism: recent

progress and controversies. European Journal of Endocrinology, 140(4), 271-286. Holloszy, J. O. (2008). Regulation by exercise of skeletal muscle content of mitochondria

and GLUT4. Journal of Physiology and Pharmacology, 59 Suppl 7, 5-18. Holt, C. L., & May, G. S. (1993). A novel phage lambda replacement Cre-lox vector that

has automatic subcloning capabilities. Gene, 133(1), 95-97. Hong, M. H., Sun, H., Jin, C. H., Chapman, M., Hu, J., Chang, W., et al. (2008). Cell-

specific activation of the human skeletal alpha-actin by androgens. Endocrinology, 149(3), 1103-1112.

Hultman, E., & Spriet, L. L. (1986). Skeletal muscle metabolism, contraction force and glycogen utilization during prolonged electrical stimulation in humans. The

Journal of Physiology, 374, 493-501.

Page 159: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

149

Hurley, B. F., Nemeth, P. M., Martin, W. H., Hagberg, J. M., Dalsky, G. P., & Holloszy, J. O. (1986). Muscle triglyceride utilization during exercise: Effect of training. The Journal of Applied Physiology, 60(2), 562-567. Huszar, D., Lynch, C. A., Fairchild-Huntress, V., Dunmore, J. H., Fang, Q., Berkemeier,

L. R., Gu, W., et al. (1997). Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell, 88(1), 131-141.

Ikeda, Y., Aihara, K., Sato, T., Akaike, M., Yoshizumi, M., Suzaki, Y., et al. (2005). Androgen receptor gene knockout male mice exhibit impaired cardiac growth and exacerbation of angiotensin II-induced cardiac fibrosis. The Journal of Biological Chemistry, 280(33), 29661-29666.

Inoue, K., Yamasaki, S., Fushiki, T., Okada, Y., & Sugimoto E. (1994). Androgen receptor antagonist suppresses exercise-induced hypertrophy of skeletal muscle. European Journal of Applied Physiology and Occupational Physiology, 69(1), 88-91. Ishikawa, T., Yamaguchi, K., Kondo, Y., Takenaka, A., & Fujisawa, M. (2008).

Metabolic syndrome in men with Klinefelter's syndrome. Urology, 71(6), 1109- 1113.

Izumiya, Y., Hopkins, T., Morris, C., Sato, K., Zeng, L., Viereck, J., et al. (2008). Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metabolism, 7(2), 159-172.

Johannsen, D. L., & Ravussin, E. (2009). The role of mitochondria in health and disease. Current Opinion in Pharmacology, 9(6), 780-786.

Jones, N. L., & Killian, K. J. (2000). Exercise limitation in health and disease. The New England Journal of Medicine, 343(9), 632-641.

Jones, R. D., Pugh, P. J., Hall, J., Channer, K. S., & Jones, T. H. (2003). Altered circulating hormone levels, endothelial function and vascular reactivity in the testicular feminised mouse. European Journal of Endocrinology, 148(1), 111-120.

Jones, M. E., Thorburn, A. W., Britt, K. L., Hewitt, K. N., Wreford, N. G., Proietto, J., Oz, O. K., et al. (2000). Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proceedings of the National Academy of Sciences of the United States of America, 97(23), 12735-12740.

Jordan, C. L., Padgett, B., Hershey, J., Prins, G., & Arnold, A. P. (1997). Ontogeny of androgen receptor immunoreactivity in lumbar motoneurons and in the sexually dimorphic levator ani muscle of male rats. The Journal of Comparative Neurology, 379(1), 88-98. Juntti, S. A., Tollkuhn, J., Wu, M. V., Fraser, E. J., Soderborg, T., Tan, S., Honda, S., et

al. (2010). The androgen receptor governs the execution, but not programming, of male sexual and territorial behaviors. Neuron, 66(2), 260-272.

Jørgensen, S. B., Treebak, J. T., Viollet, B., Schjerling, P., Vaulont, S., Wojtaszewski, J. F. P., & Richter, E. A. (2007). Role of AMPKalpha2 in basal, training-, and AICAR-induced GLUT4, hexokinase II, and mitochondrial protein expression in mouse muscle. American Journal of Physiology. Endocrinology and Metabolism, 292(1), E331-339.

Kadi, F. (2008). Cellular and molecular mechanisms responsible for the action of testosterone on human skeletal muscle. A basis for illegal performance enhancement. British Journal of Pharmacology, 154(3), 522-528.

Page 160: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

150

Kadi, F., Bonnerud, P., Eriksson, A., & Thornell, L. E. (2000). The expression of androgen receptors in human neck and limb muscles: effects of training and self- administration of androgenic-anabolic steroids. Histochemistry and Cell Biology, 113(1), 25-29. Kadi, F., Eriksson, A., Holmner, S., & Thornell, L. E. (1999). Effects of anabolic steroids on the muscle cells of strength-trained athletes. Medicine and Science in Sport and Exercise, 31(11), 1528-1534. Kammoun, H. L., Chabanon, H., Hainault, I., Luquet, S., Magnan, C., Koike, T., et al.

(2009). GRP78 expression inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in mice. The Journal of Clinical Investigation, 119(5), 1201-1215.

Kahaly, G. J. (2010). The thyrocyte-fibrocyte link: closing the loop in the pathogenesis of Graves’ disease? The Journal of Clinical Endocrinology and Metabolism, 95, 62-

65. Kawano, H., Sato, T., Yamada, T., Matsumoto, T., Sekine, K., Watanabe, T., et al.

(2003). Suppressive function of androgen receptor in bone resorption. Proceedings of the National Academy of Sciences of the United States of America, 100(16), 9416-9421.

Kee, A. J., Gunning, P. W., & Hardeman, E. C. (2009). Diverse roles of the actin cytoskeleton in striated muscle. Journal of Muscle Research and Cell Motility, 30(5-6), 187-197.

Kennedy, W. R., Alter, M., & Sung, J. H. (1968). Progressive proximal spinal and bulbar muscular atrophy of late onset. A sex-linked recessive trait. Neurology, 18(7), 671-680.

Kerkhofs, S., Denayer, S., Haelens, A., & Claessens, F. (2009). Androgen receptor knockout and knock-in mouse models. Journal of Molecular Endocrinology, 42(1), 11-17.

Kerr, J. E., Allore, R. J., Beck, S. G., & Handa, R. J. (1995). Distribution and hormonal regulation of androgen receptor (AR) and AR messenger ribonucleic acid in the rat hippocampus. Endocrinology, 136(8), 3213-3221.

Klivenyi, P., Ferrante, R. J., Matthews, R. T., Bogdanov, M. B., Klein, A. M., Andreassen, O. A., Mueller, G., et al. (1999). Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nature Medicine, 5(3), 347-350.

Klover, P., Chen, W., Zhu, B. M., Hennighausen, L. (2009). Skeletal muscle growth and fiber composition in mice are regulated through the transcription factors STATa5/b: linking growth hormone to the androgen receptor. The FASEB

Journal, 23(9), 3140-3148. Koopman, P., Gubbay, J., Vivian, N., Goodfellow, P., & Lovell-Badge, R. (1991). Male

development of chromosomally female mice transgenic for Sry. Nature, 351(6322), 117-121.

Krude, H., Biebermann, H., Luck, W., Horn, R., Brabant, G., & Grüters, A. (1998). Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nature Genetics, 19(2), 155-157.

Kurz, E. M., Cover, A. R., & Sengelaub, D. R. (1992). Testosterone fails to save androgen-sensitive rat motoneurons following early target removal. Brain

Page 161: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

151

Research. Developmental Brain Research, 70(2), 181-189. La Spada, A. R., Roling, D. B., Harding, A. E., Warner, C. L., Spiegel, R., Hausmanowa-

Petrusewicz, I., Yee, W. C., et al. (1992). Meiotic stability and genotype- phenotype correlation of the trinucleotide repeat in X-linked spinal and bulbar muscular atrophy. Nature Genetics, 2(4), 301-304.

La Spada, A. R., Wilson, E. M., Lubahn, D. B., Harding, A. E., & Fischbeck, K. H. (1991). Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature, 352(6330), 77-79.

Laaksonen, D. E., Niskanen, L., Punnonen, K., Nyyssönen, K., Tuomainen, T., Valkonen, V., & Salonen, J. T. (2005). The metabolic syndrome and smoking in relation to hypogonadism in middle-aged men: a prospective cohort study. The Journal of Clinical Endocrinology and Metabolism, 90(2), 712-719.

Lai, J., Lai, K., Chuang, K., Chang, P., Yu, I., Lin, W., et al. (2009). Monocyte/macrophage androgen receptor suppresses cutaneous wound healing in mice by enhancing local TNF-alpha expression. The Journal of Clinical Investigation, 119(12), 3739-3751.

Lee, D. K., & Chang, C. (2003). Endocrine mechanisms of disease: Expression and degradation of androgen receptor: mechanism and clinical implication. The Journal of Clinical Endocrinology and Metabolism, 88(9), 4043-4054.

Lefterova, M. I., & Lazar, M. A. (2009). New developments in adipogenesis. Trends in Endocrinology and Metabolism, 20(3), 107-114.

Lehman, J. J., Barger, P. M., Kovacs, A., Saffitz, J., Medeiros, D.M., & Kelley, D. P. (2000). Peroxisome proliferator-activated receptor γ coactivator-1 promotes cardiac mitochondrial biogenesis. The Journal of Clinical Investigation, 106(7), 847-856. Lin, J., Handschin, C., & Spiegelman, B. M. (2005). Metabolic control through the PGC-1 family of transcription coactivators. Cell Metabolism, 1(6), 361-370. Lin, J., Wu, P., Tarr, P. T., Lindenberg, K. S., St-Pierre, J., Zhang, C., Mootha, V. K., et

al. (2004). Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell, 119(1), 121-135.

Lin, J., Wu, H., Tarr, P. T., Zhang, C., Wu, Z., Boss, O., Michael, L. F., et al. (2002). Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature, 418(6899), 797-801.

Lin, H., Xu, Q., Yeh, S., Wang, R., Sparks, J. D., & Chang, C. (2005). Insulin and leptin resistance with hyperleptinemia in mice lacking androgen receptor. Diabetes, 54(6), 1717-1725.

Lin, H., Yu, I., Wang, R., Chen, Y., Liu, N., Altuwaijri, S., et al. (2008). Increased hepatic steatosis and insulin resistance in mice lacking hepatic androgen receptor. Hepatology, 47(6), 1924-1935.

Lönnqvist, F., Thörne, A., Large, V., & Arner, P. (1997). Sex differences in visceral fat lipolysis and metabolic complications of obesity. Arteriosclerosis, Thrombosis, and Vascular Biology, 17(7), 1472-1480.

Lowell, B. B., & Flier, J. S. (1997). Brown adipose tissue, beta 3-adrenergic receptors, and obesity. Annual Review of Medicine, 48, 307-316.

Lubahn, D. B., Joseph, D. R., Sar, M., Tan, J., Higgs, H. N., Larson, R. E., French, F. S., et al. (1988). The human androgen receptor: complementary deoxyribonucleic

Page 162: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

152

acid cloning, sequence analysis and gene expression in prostate. Molecular Endocrinology, 2(12), 1265-1275.

Luczak, E. D., & Leinwand, L. A. (2009). Sex-based cardiac physiology. Annual Review of Physiology, 71, 1-18.

Luisetto, G., & Camozzi, V. (2009). Statins, fracture risk, and bone remodeling. Journal of Endocrinological Investigation, 32(4 Suppl), 32-37.

Lutter, M., Sakata, I., Osborne-Lawrence, S., Rovinsky, S. A., Anderson, J. G., Jung, S., et al. (2008). The orexigenic hormone ghrelin defends against depressive

symptoms of chronic stress. Nature Neuroscience, 11(7), 2008. Lyon, M. F., & Hawkes, S. G. (1970). X-linked gene for testicular feminization in the

mouse. Nature, 227(5264), 1217-1219. MacDougald, O. A., & Mandrup, S. (2002). Adipogenesis: forces that tip the scales.

Trends in Endocrinology and Metabolism, 13(1), 5-11. MacLean, H. E., Chiu, W. S. M., Notini, A. J., Axell, A., Davey, R. A., McManus, J. F.,

et al. (2008). Impaired skeletal muscle development and function in male, but not female, genomic androgen receptor knockout mice. The FASEB Journal, 22(8), 2676-2689.

MacLean, H. E., & Handelsman, D. J. (2009). Unraveling androgen action in muscle: genetic tools probing cellular mechanisms. Endocrinology, 150(8), 3437-3439.

Malina, R. M. (1969). Quantification of fat, muscle and bone in man. Clinical Orthopaedics and Related Research, 65, 9-38.

Mänttäri, S., Anttila, K., & Järvilehto, M. (2008). Testosterone stimulates myoglobin expression in different muscles of the mouse. Journal of Comparative Physiology. B, Biochemical, Systemic, and Environmental Physiology, 178(7), 899-907.

Mantzoros, C. S., & Flier, J. S. (2000). Editorial: leptin as a therapeutic agent--trials and tribulations. The Journal of Clinical Endocrinology and Metabolism, 85(11), 4000-4002.

Mårin, P., Lönn, L., Andersson, B., Odén, B., Olbe, L., Bengtsson, B. A., & Björntorp, P. (1996). Assimilation of triglycerides in subcutaneous and intraabdominal adipose tissues in vivo in men: effects of testosterone. The Journal of Clinical Endocrinology and Metabolism, 81(3), 1018-1022.

Marsh, J. D., Lehmann, M. H., Ritchie, R. H., Gwathmey, J. K., Green, G. E., & Schiebinger, R. J. (1998). Androgen receptors mediate hypertrophy in cardiac myocytes. Circulation, 98(3), 256-261.

Matsumoto, T., Takeyama, K., Sato, T., & Kato, S. (2003). Androgen receptor functions from reverse genetic models. The Journal of Steroid Biochemistry and Molecular Biology, 85(2-5), 95-99.

Mauro, A. (1961). Satellite cell of skeletal muscle fibers. The Journal of Biophysical and Biochemical Cytology, 9, 493-495. Mayes, J. S., & Watson, G. H. (2004). Direct effects of sex steroid hormones on adipose

tissues and obesity. Obesity Reviews, 5(4), 197-216. McEwen, B. S. (1992). Steroid hormones: effect on brain development and function. Hormone Research, 37(3), 1-10. McGee, S. L., & Hargreaves, M. (2010). AMPK-mediated regulation of transcription in

skeletal muscle. Clinical Science, 118(8), 507-518. McKenna, K. E. & Nadelhaft, I. (1986). The organization of the pudendal nerve in the

Page 163: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

153

male and female rat. The Journal of Comparative Neurology, 248(4), 532-549. Menzies, F. M., Ince, P. G., & Shaw, P. J. (2002). Mitochondrial involvement in

amyotrophic lateral sclerosis. Neurochemistry International, 40(6), 543-551. Merrill, G. F., Kurth, E. J., Hardie, D. G., & Winder, W. W. (1997). AICA riboside increases AMP-activated protein kinase, fatty acid oxidation, and glucose uptake in rat muscle. American Journal of Physiology, 273(6 Pt 1), E1107-E1112. Merry, D. E., Kobayashi, Y., Bailey, C. K., Taye, A. A., & Fischbeck, K. H. (1998).

Cleavage, aggregation and toxicity of the expanded androgen receptor in spinal and bulbar muscular atrophy. Human Molecular Genetics, 7(4), 693-701.

Miniou, P., Tiziano, D., Frugier, T., Roblot, N., Le Meur, M., & Melki, J. (1999). Gene targeting restricted to mouse striated muscle lineage. Nucleic Acids Research, 27(19), e27.

Minnich, A., Tian, N., Byan, L., & Bilder, G. (2001). A potent PPARalpha agonist stimulates mitochondrial fatty acid beta-oxidation in liver and skeletal muscle. American Journal of Physiology. Endocrinology and Metabolism, 280(2), E270- 279.

Monks, D. A., Getsios, S., MacCalman, C. D., & Watson, N. V. (2001a). N-cadherin is regulated by gonadal steroids in adult sexually dimorphic spinal motoneurons. Journal of Neurobiology, 47(4), 255-264.

Monks, D. A., Getsios, S., MacCalman, C. D., & Watson, N. V. (2001b). N-cadherin is regulated by gonadal steroids in the adult hippocampus. Proceedings of the National Academy of Sciences of the United States of America, 98(3), 1312-1316.

Monks, D. A., Johansen, J. A., Mo, K., Rao, P., Eagleson, B., Yu, Z., et al. (2007). Overexpression of wild-type androgen receptor in muscle recapitulates polyglutamine disease. Proceedings of the National Academy of Sciences of the United States of America, 104(46), 18259-18264.

Monks, D. A., Kopachik, W., Breedlove, S. M., & Jordan, C. L. (2006). Anabolic r esponsiveness of skeletal muscles correlates with androgen receptor protein but not mRNA. Canadian Journal of Physiology and Pharmacology, 84(2), 273-277.

Monks, D. A., O'Bryant, E. L., & Jordan, C. L. (2004). Androgen receptor immunoreactivity in skeletal muscle: enrichment at the neuromuscular junction. The Journal of Comparative Neurology, 473(1), 59-72.

Monks, D. A., Rao, P., Mo, K., Johansen, J. A., Lewis, G., & Kemp, M. Q. (2008). Androgen receptor and Kennedy disease/spinal bulbar muscular atrophy. Hormones and Behavior, 53(5), 729-740.

Monks, D. A., Vanston, C. M., & Watson, N. V. (1999). Direct androgenic regulation of calcitonin gene-related peptide expression in motoneurons of rats with mosaic androgen insensitivity. The Journal of Neuroscience, 19(13), 5597-5601.

Monks, D. A., & Watson, N. V. (2001). N-cadherin expression in motoneurons is directly regulated by androgens: a genetic mosaic analysis in rats. Brain Research, 895(1- 2), 73-79.

Mootha, V. K., Lindgren, C. M., Eriksson, K., Subramanian, A., Sihag, S., Lehar, J., Puigserver, P., et al. (2003). PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genetics, 34(3), 267-273.

Page 164: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

154

Morris, J. A., Jordan, C. L., & Breedlove, S. M. (2004). Sexual differentiation of the vertebrate nervous system. Nature Neuroscience, 7(10), 1034-1039.

Morris, J. A., Jordan, C. L., Dugger, B. N., & Breedlove, S. M. (2005). Partial demasculinization of several brain regions in adult male (XY) rats with a dysfunctional androgen receptor gene. The Journal of Comparative Neurology, 487(2), 217-226.

Movérare-Skrtic, S., Venken, K., Andersson, N., Lindberg, M. K., Svensson, J., Swanson, C., et al. (2006). Dihydrotestosterone treatment results in obesity and altered lipid metabolism in orchidectomized mice. Obesity, 14(4), 662-672.

Mujica, V., Leiva, E., Icaza, G., Diaz, N., Arredondo, M., Moore-Carrasco, R., Orrego, R., et al. (2008). Evaluation of metabolic syndrome in adults of Talca city, Chile. Nutrition Journal, 7, 14.

Munday, M. R., Carling, D., and Hardie, D. G. (1988). Negative interactions between phosphorylation of acetyl-CoA carboxylase by the cyclic AMP-dependent and AMP-activated protein kinases. FEBS Letters, 223(1-2), 217-222. Musatov, S., Chen, W., Pfaff, D. W., Mobbs, C. V., Yang, X., Clegg, D. J., Kaplitt, M.

G., et al. (2007). Silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. Proceedings of the National Academy of Sciences of the United States of America, 104(7), 2501-2506.

Naess, O., Haug, E., Attramadal, A., Aakvaag A., Hansson, V., & French, F. (1976). Androgen receptors in the anterior pituitary and central nervous system of the androgen “insensitive” (Tfm) rat: correlation between receptor binding and effects of androgens on gonadotropin secretion. Endocrinology, 99(4), 1295-1303.

Nakazato M., Murakami, N., Date, Y., Kojima, M., Matsuo, H., Kangawa K., et al. (2001). A role for ghrelin in the central regulation of feeding. Nature, 409(6817), 194-198.

Narkar, V. A., Downes, M., Yu, R. T., Embler, E., Wang, Y., Banayo, E., et al. (2008). AMPK and PPARδ agonists are exercise mimetics. Cell, 134(3), 405-415.

Niel, L., Shah, A. H., Lewis, G. A., Mo, K., Chatterjee, D., Fernando, S. M., et al. (2009). Sexual differentiation of the spinal nucleus of the bulbocavernosus is not mediated solely by androgen receptors in muscle fibers. Endocrinology, 150(7), 3207-3213.

Niel, L., Willemsen, K. R., Volante, S. N., & Monks, D. A. (2008). Sexual dimorphism and androgen regulation of satellite cell population in differentiating rat levator ani muscle. Developmental Neurobiology, 68(1), 115-122.

Nordeen, E. J., Nordeen, K. W., Sengelaub, D. R., & Arnold, A. P. (1985). Androgens prevent normally occurring cell death in a sexually dimorphic spinal nucleus. Science, 229(4714), 671-673. Olney, R. C. (2009). Mechanisms of impaired growth: effect of steroids on bone and

cartilage. Hormone Research, 72 Suppl 1, 30-35. Orallo, F. (1996). Regulation of cytosolic calcium levels in vascular smooth muscle. Pharmacology and Therapeutics, 69(3), 153-171. Ophoff, J., Callewaert, F., Venken, K., De Gendt, K., Ohlsson, C., Gayan-Ramirez, G., et

al. (2009). Physical activity in the androgen receptor knockout mouse: evidence for reversal of androgen deficiency on cancellous bone. Biochemical and Biophysical Research Communications, 378(1), 139-144.

Page 165: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

155

Ophoff, J., Van Proeyen, K., Callewaert, F., De Gendt, K., De Bock, K., Vanden Bosch, A., et al. (2009). Androgen signaling in myocytes contributes to the maintenance of muscle mass and fiber type regulation but not to muscle strength or fatigue. Endocrinology, 150(8), 3558-3566.

Page, S. T., Marck, B. T., Tolliver, J. M., & Matsumoto, A. M. (2008). Tissue selectivity of the anabolic steroid, 19-nor-4-androstenediol-3beta,17beta-diol in male Sprague Dawley rats: selective stimulation of muscle mass and bone mineral density relative to prostate mass. Endocrinology, 149(4), 1987-1993.

Pedersen, B. K., Akerström, T. C. A., Nielsen, A. R., & Fischer, C. P. (2007). Role of myokines in exercise and metabolism. Journal of Applied Physiology, 103(3), 1093-1098.

Pedersen, B. K., & Febbraio, M. (2005). Muscle-derived interleukin-6--a possible link between skeletal muscle, adipose tissue, liver, and brain. Brain, Behavior, and Immunity, 19(5), 371-376.

Petersen, K. F., Dufour, S., Befroy, D., Garcia, R., & Shulman, G. I. (2004). Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. The New England Journal of Medicine, 350(7), 664-671.

Pohl, J., Ring, A., Korkmaz, U., Ehehalt, R., & Stremmel, W. (2005). FAT/CD36- mediated long-chain fatty acid uptake in adipocytes requires plasma membrane rafts. Molecular Biology of the Cell, 16(1), 24-31.

Powers, M. L. & Florini, J. R. (1975). A direct effect of testosterone on muscle cells in tissue culture. Endocrinology, 97(4), 1043-1047. Psarra, A. G., & Sekeris, C. E. (2008). Steroid and thyroid hormone receptors in

mitochondria. IUBMB Life, 60(4), 210-223. Psarra, A. M. G., Solakidi, S., & Sekeris, C. E. (2006). The mitochondrion as a primary

site of action of regulatory agents involved in neuroimmunomodulation. Annals of the New York Academy of Sciences, 1088, 12-22.

Rao, P., & Monks, D. A. (2009). A tetracycline-inducible and skeletal muscle-specific Cre recombinase transgenic mouse. Developmental Neurobiology, 69(6), 401-406.

Ranganathan, S., Harmison, G. G., Meyertholen, K., Pennuto, M., Burnett, B. G., & Fischbeck, K. H. (2009). Mitochondrial abnormalities in spinal and bulbar muscular atrophy. Human Molecular Genetics, 18(1), 27-42.

Riggs, B. L., Khosla, S., & Melton, L. J. (2002). Sex steroids and the construction and conservation of the adult skeleton. Endocrine Reviews, 23(3), 279-302.

Ritov, V. B., Menshikova, E. V., Azuma, K., Wood, R. J., Toledo, F. G. S., Goodpaster, B. H., Ruderman, N. B., et al. (2009). Deficiency of Electron Transport Chain in Human Skeletal Muscle Mitochondria in Type 2 Diabetes Mellitus and Obesity. American Journal of Physiology. Endocrinology and Metabolism. (In Press).

Ritov, V. B., Menshikova, E. V., He, J., Ferrell, R. E., Goodpaster, B. H., & Kelley, D. E. (2005). Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes, 54(1), 8-14.

Roche, P. J., Hoare, S. A., & Parker, M. G. (1992). A consensus DNA-binding site for the androgen receptor. Molecular Endocrinology, 6(12), 2229-2235.

Roselli C. E., Salisbury, R. L., & Resko, J. A. (1987). Genetic evidence for androgen- dependent and independent control of aromatase activity in the rat brain Endocrinology, 128(3), 2205-2210.

Page 166: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

156

Rosen, E. D., & Spiegelman, B. M. (2000). Molecular regulation of adipogenesis. Annual Review of Cell and Developmental Biology, 16, 145-171.

Rosenfeld, J. M., Daley, J. D., Ohno, S., & YoungLai, E. V. (1977). Central aromatization of estrogen of testosterone in testicular feminized mice. Experientia, 33(10), 1392-1393.

Ruizeveld de Winter, J. A., Trapman, J., Vermey, M., Mulder, E., Zegers, N. D., & van der Kwast, T. H. (1991). Androgen receptor expression in human tissues: an immunohistochemical study. The Journal of Histochemistry and Cytochemistry: Official Journal of the Histochemistry Society, 39(7), 927-936.

Schmidt, A., Harada, S., Kimmel, D. B., Bai, C., Chen, F., Rutledge, S. J., Vogel, R. L., et al. (2009). Identification of anabolic selective androgen receptor modulators with reduced activities in reproductive tissues and sebaceous glands. The Journal of Biological Chemistry, 284(52), 36367-36376.

Schwartz, M. W., Woods, S. C., Porte, D., Seeley, R. J., & Baskin, D. G. (2000). Central nervous system control of food intake. Nature, 404(6778), 661-671.

Scott, W., Stevens, J., & Binder-Macleod, S. A. (2001). Human skeletal muscle fiber type classifications. Physical Therapy, 81(11), 1810-1816. Seidell, J. C., Björntorp, P., Sjöström, L., Kvist, H., & Sannerstedt, R. (1990). Visceral

fat accumulation in men is positively associated with insulin, glucose, and C- peptide levels, but negatively with testosterone levels. Metabolism: Clinical and Experimental, 39(9), 897-901.

Sengelaub, D. R., & Forger, N. G. (2008). The spinal nucleus of the bulbocavernosus: firsts in androgen-dependent neural sex differences. Hormones and Behavior, 53(5), 596-612.

Shaffer, P. L., Jivan, A., Dollins, D. E., Claessens, F., & Gewirth, D. T. (2004). Structural basis of androgen receptor binding to selective androgen response elements. Proceedings of the National Academy of Sciences of the United States of America, 101(14), 4758-4763.

Shi, H., Seeley, R. J., & Clegg, D. J. (2009). Sexual differences in the control of energy homeostasis. Frontiers in Neuroendocrinology, 30(3), 396-404.

Simerly, R. B. (2002). Wired for reproduction: organization and development of sexually dimorphic circuits in the mammalian forebrain. Annual Review of Neuroscience, 25, 507-536.

Simpson, E. R., & Davis, S. R. (2000). Another role highlighted for estrogens in the male: sexual behavior. Proceedings of the National Academy of Sciences of the United States of America, 97(26), 14038-14040.

Sinclair, A. H., Berta, P., Palmer, M. S., Hawkins, J. R., Griffiths, B. L., Smith, M. J., Foster, J. W., et al. (1990). A gene from the human sex-determining region encodes a protein with homology to a conserved DNA-binding motif. Nature, 346(6281), 240-244.

Singh, R., Bhasin, S., Braga, M., Artaza, J. N., Pervin, S., Taylor, W. E., et al. (2009). Regulation of myogenic differentiation by androgens: cross talk between androgen receptor/ beta-catenin and follistatin/transforming growth factor-beta signaling pathways. Endocrinology, 150(3), 1259-1268.

Singh, R., Artaza, J. N., Taylor, W. E., Braga, M., Yuan, X., Gonzalez-Cadavid, N. F., et al. (2006). Testosterone inhibits adipogenic differentiation in 3T3-L1 cells:

Page 167: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

157

nuclear translocation of androgen receptor complex with beta-catenin and T-cell factor 4 may bypass canonical Wnt signaling to down-regulate adipogenic transcription factors. Endocrinology, 147(1), 141-154.

Singh, R., Artaza, J. N., Taylor, W. E., Gonzalez-Cadavid, N. F., & Bhasin, S. (2003). Androgens stimulate myogenic differentiation and inhibit adipogenesis in C3H 10T1/2 pluripotent cells through an androgen receptor-mediated pathway. Endocrinology, 144(11), 5081-5088.

Sinha-Hikim, I., Artaza, J., Woodhouse, L., Gonzalez-Cadavid, N., Singh, A. B., Lee, M. I., Storer, T. W., et al. (2002). Testosterone-induced increase in muscle size in healthy young men is associated with muscle fiber hypertrophy. American Journal of Physiology. Endocrinology and Metabolism, 283(1), E154-164.

Sinha-Hikim, L., Roth, S. M., Lee, M. I., & Bhasin, S. (2003). Testosterone-induced muscle hypertrophy is associated with an increase in satellite cell number in healthy, young men. American Journal of Physiology. Endocrinology and Metabolism, 285(1), E197-205. Sinha-Hikim, L., Taylor, W. E., Gonzalez-Cadavid N. F., Zheng, W., & Bhasin, S. (2004). Androgen receptor in human skeletal muscle and cultured muscle satellite cells: up-regulation by androgen treatment. The Journal of Clinical Endocrinology and Metabolism, 89(10), 5245-5255. Snyder, P. J., Peachey, H., Berlin, J. A., Hannoush, P., Haddad, G., Dlewati, A., et al.

(2000). Effects of testosterone replacement in hypogonadal men. The Journal of Clinical Endocrinology and Metabolism, 85(8), 2670-2677.

Solakidi, S., Psarra, A. G., Nikolaropoulos, S., & Sekeris, C. E. (2005). Estrogen receptors alpha and beta (ERalpha and ERbeta) and androgen receptor (AR) in human sperm: localization of ERbeta and AR in mitochondria of the midpiece. Human Reproduction, 20(12), 3481-3487.

Sopher, B. L., Thomas, P. S., LaFevre-Bernt, M. A., Holm, I. E., Wilke, S. A., Ware, C. B., Jin, L., et al. (2004). Androgen receptor YAC transgenic mice recapitulate SBMA motor neuronopathy and implicate VEGF164 in the motor neuron degeneration. Neuron, 41(5), 687-699.

Spiegelman, B. M., & Flier, J. S. (2001). Obesity and the regulation of energy balance. Cell, 104(4), 531-543.

Stanley, B. G., Kyrkouli, S. E., Lampert, S., & Leibowitz, S. F. (1986). Neuropeptide Y chronically injected into the hypothalamus: a powerful neurochemical inducer of hyperphagia and obesity. Peptides, 7(6), 1189-1192.

Staron, R. S. (1997). Human skeletal muscle fiber types: delineation, development, and distribution. Canadian Journal of Applied Physiology, 22(4), 307-327. Steinberg, G. R., Smith, A. C., van Denderen, B. J. W., Chen, Z. P., Murthy, S., Campbell, D. J., et al. (2004). AMP-activated protein kinase is not down- regulated in human skeletal muscle of obese females. The Journal of Clinical Endocrinology and Metabolism, 89(9), 4575-4580. Stoppani, J., Hildebrandt, A. L., Sakamoto, K., Cameron-Smith, D., Goodyear, L. J., &

Neufer, P. D. (2002). AMP-activated protein kinase activates transcription of the UCP3 and HKII genes in rat skeletal muscle. American Journal of Physiology. Endocrinology and Metabolism, 283(6), E1239-1248.

Page 168: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

158

Svensson, J., Movérare-Skrtic, S., Windahl, S., Swanson, C., & Sjögren, K. (2010). Both estrogen and androgen receptor stimulation maintains skeletal muscle mass in gonadectomized male mice but mainly via different pathways. Journal of Molecular Endocrinology. (In Press).

Tamauchi, T., Kamon, J., Minokoshi, Y., Ito, Y., Waki, H., Uchida, S., et al. (2002). Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nature Medicine, 8(11), 1288-1295. Tedesco, F. S., Dellavalle, A., Diaz-Manera, J., Messina, G., & Cossu, G. (2010). Repairing skeletal muscle: regenerative potential of skeletal muscle stem cells. The Journal of Clinical Investigation, 120(1), 11-19. Ting, H., & Chang, C. (2008). Actin associated proteins function as androgen receptor

coregulators: an implication of androgen receptor's roles in skeletal muscle. The Journal of Steroid Biochemistry and Molecular Biology, 111(3-5), 157-163.

Tobet, S. A., Bless, E. P., & Schwarting, G. A. (2001). Developmental aspect of the gonadotropin-releasing hormone system. Molecular and Cellular Endocrinology, 185(1-2), 173-184.

Trapman, J., Ris-Stalpers, C., van der Korput, J. A., Kuiper, G. G., Faber, P. W., Romijn, J. C., Mulder, E., et al. (1990). The androgen receptor: functional structure and expression in transplanted human prostate tumors and prostate tumor cell lines. The Journal of Steroid Biochemistry and Molecular Biology, 37(6), 837-842.

Tschop, M., Smiley, D. L., & Heiman, M. L. (2000). Ghrelin induces adiposity in rodents. Nature, 407(6806), 908-913.

Turcotte, L. P., Swenberger, J. R., Zavitz Tucker, M., & Yee, A. J. (2001). Increased fatty acid uptake and altered fatty acid metabolism in insulin-resistant muscle of obese Zucker rats. Diabetes, 51(6), 1477-1482. Turner, N., Bruce, C. R., Beale, S. M., Hoehn, K. L., So, T., Rolph, M. S., & Cooney, G.

J. (2007). Excess lipid availability increases mitochondrial fatty acid oxidative capacity in muscle: evidence against a role for reduced fatty acid oxidation in lipid-induced insulin resistance in rodents. Diabetes, 56(8), 2085-2092.

Umetani, M., Domoto, H., Gormley, A. K., Yuhanna, I. S., Cummins, C. L., Javitt, N. B., Korach, K. S., et al. (2007). 27-Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nature Medicine, 13(10), 1185- 1192.

Vanderschueren, D., Van Herck, E., Suiker, A. M., Visser, W. J., Schot, L. P., Chung, K., Lucas, R. S., et al. (1993). Bone and mineral metabolism in the androgen-resistant (testicular feminized) male rat. Journal of Bone and Mineral Research, 8(7), 801- 809.

Vanderschueren, D., Van Herck, E., Geusens, P., Suiker, A., Visser, W., Chung, K., & Bouillon, R. (1994). Androgen resistance and deficiency have different effects on the growing skeleton of the rat. Calcified Tissue International, 55(3), 198-203.

Vanderschueren, D., Van Herck, E., Nijs, J., Ederveen, A. G., De Coster, R., & Bouillon, R. (1997). Aromatase inhibition impairs skeletal modeling and decreases bone mineral density in growing male rats. Endocrinology, 138(6), 2301-2307.

Walsh, P. C. (2010). Chemoprevention of prostate cancer. The New England Journal of Medicine, 362(13), 1237-1238

Page 169: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

159

Wang, J., Liu, R., Hawkins, M., Barzilai, N., & Rossetti, L. (1998). A nutrient-sensing pathway regulates leptin gene expression in muscle and fat. Nature, 393(6686), 684-688.

Wang, Y., Zhang, C., Yu, R. T., Cho, H. K., Nelson, M. C., Bayuga-Ocampo, C. R., et al. (2004). Regulation of muscle fiber type and running endurance by peroxisome-proliferator-activated receptor delta. PLoS Biology, 2(10), e294. Wang, Y., Lee, C., Tiep, S., Yu, R. T., Ham, J., Kang, H., et al. (2003). Peroxisome-

proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell, 113(2), 159-170.

Weber, K., Brück, P., Mikes, Z., Küpper, J., Klingenspor, M., & Wiesner, R. J. (2002). Glucocorticoid hormone stimulates mitochondrial biogenesis specifically in skeletal muscle. Endocrinology, 143(1), 177-184.

Wehling, M. (1997). Specific, nongenomic actions of steroid hormones. Annual Review of Physiology, 59, 365-393.

Welle, S., Tawil, R., & Thornton, C. A. (2008). Sex-related differences in gene expression in human skeletal muscle. PLoS One, 3(1), e1385.

Wende, A. R., Schaeffer, P. J., Parker, G. J., Zechner, C., Han, D. H., Chen, M. M., et al. (2007). A role for the transcriptional coactivator PGC-1α in muscle refueling.

The Journal of Biological Chemistry, 282(50), 36642-36651. Wenz, T., Diaz, F., Spiegelman, B. M., & Moraes, C. T. (2008). Activation of the

PPAR/PGC-1alpha pathway prevents a bioenergetic deficit and effectively improves a mitochondrial myopathy phenotype. Cell Metabolism, 8(3), 249-256.

Wenz, T., Rossi, S. G., Rotundo, R. L., Spiegelman, B. M., & Moraes, C. T. (2009). Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging. Proceedings of the National Academy of Sciences of the United States of America, 106(48), 20405-20410.

Williams, G. A., Wang, Y., Callon, K. E., Watson, M., Lin, J., Lam, J. B. B., Costa, J. L., et al. (2009). In vitro and in vivo effects of adiponectin on bone. Endocrinology, 150(8), 3603-3610.

Wilson, J. D. (1988). Androgen abuse by athletes. Endocrine Reviews, 9(2), 181-199. Wilson, J. D. (1999). The role of androgens in male gender role behavior. Endocrine

Reviews, 20(5), 726-737. Wiren, K. M., Zhang, X., Chang, C., Keenan, E., & Orwoll, E. S. (1997). Transcriptional

up-regulation of the human androgen receptor by androgen in bone cells. Endocrinology, 138(6), 2291-2300.

Wu Z., Puigserver, P., Andersson, U., Zhang, C., Adelmant, G., Mootha, V., et al. (1999). Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell, 98(1), 115-124.

Yanase, T., Fan, W., Kyoya, K., Min, L., Takayanagi, R., Kato, S., et al. (2008). Androgens and metabolic syndrome: lessons from androgen receptor knock out (ARKO) mice. The Journal of Steroid Biochemistry and Molecular Biology, 109(3-5), 254-257.

Yang, S., Liu, R., Perez, E. J., Wen, Y., Stevens, S. M., Valencia, T., Brun-Zinkernagel, A., et al. (2004). Mitochondrial localization of estrogen receptor beta. Proceedings of the National Academy of Sciences of the United States of America, 101(12), 4130-4135.

Page 170: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

160

Yarbrough, W. G., Quarmby, V. E., Simental, J. A., Joseph, D. R., Sar, M., Lubahn, D. B., et al. (1990). A single base mutation in the androgen receptor gene causes androgen insensitivity in the testicular feminized rat. The Journal of Biological Chemistry, 265(15), 8893-8900.

Yeh, S., Tsai, M., Xu, Q., Mu, X., Lardy, H., Huang, K., et al. (2002). Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proceedings of the National Academy of Sciences of the United States of America, 99(21), 13498- 13503.

Yu, Z., Dadgar, N., Albertelli, M., Gruis, K., Jordan, C., Robins, D. M., & Lieberman, A. P. (2006). Androgen-dependent pathology demonstrates myopathic contribution to the Kennedy disease phenotype in a mouse knock-in model. The Journal of Clinical Investigation, 116(10), 2663-2672.

Yu, I., Lin, H., Liu, N., Wang, R., Sparks, J. D., Yeh, S., et al. (2008). Hyperleptinemia without obesity in male mice lacking androgen receptor in adipose tissue. Endocrinology, 149(5), 2361-2368.

Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L., & Friedman, J. M. (1994). Positional cloning of the mouse obese gene and its human homologue. Nature, 372(6505), 425-432.

Zitzmann, M. (2009). Testosterone deficiency, insulin resistance and the metabolic syndrome. Nature Reviews. Endocrinology, 5(12), 673-681.

Zuloaga, D. G., Puts, D. A., Jordan, C. L., & Breedlove, S. M. (2008). The role of androgen receptors in the masculinization of brain and behavior: what we've learned from the testicular feminization mutation. Hormones and Behavior, 53(5), 613-626.

Page 171: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

161

Tables and Figures

TABLE 1: Summary of Animals Used

Experiment WT M

Rats

Tg M

Rats

Tfm M

Rats

Tfm /Tg M

Rats

WT F

Rats

Tg F

Rats

Tfm F

Rats

Tfm /Tg F

Rats

WT M

Mice

L78 M

Mice

Exp. I

RT-PCR 3 3

Exp. 2

Body Composition 11 11 7 9 8 9 7 7 5 6

Dissections 10 12 7 10

Exp. 3

Testosterone Treatment 8 9

Exp. 4

Adipose Histology 6 6 6 4

Exp. 5

RMR 10 12 7 10 5 6

Activity Measures 4 6 3 5

Page 172: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

162

TABLE 2: Primer Sequences

Application Gene Name (Species) Sequenece PCR Human AR (Rat) F: GGACAGGGCACTACCGAG

R: GGCTGAATCTTCCACCTAC PCR Rat AR (Rat) F: GCAACTTGCATGTGGATGA

R: TGAAAACCAGGTCAGGTGC PCR GAPDH (Rat) F: ATGGGAAGCTGGTCATCAAC

R: GGATGCAGGGATGATGTTCT PCR Rat AR (Mouse) F: AGTAGCCAACAGGGAAGGGT

R: GAGGCAGCCGCTCTCAGGGTG PCR TSH (Mouse) F: AACGGAGAGTGGGTCATCAC

R: CATTGGGTTAAGCACACAGG Endpoint PCR Human AR (Rat) F: AGGAAGCAGTATCCGAAGGCA

R: GGACACCGACACTGCCTTACA

Page 173: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

163

Figure 1: Characterization of Transgene Expression. RT-PCR reveals expression of the transgene in urinary bladder, heart and skeletal muscle of Tg animals only.

Page 174: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

164

Figure 2: Transgene Expression Regulates Body Composition in Males. Representative DXA images from WT (A), HSA-AR (B), Tfm (C), and HSA-AR/Tfm (D) male rats are shown. Note the reduced mass in the abdominal region of Tg animals. (E) HSA-AR has no effect on body mass, but this parameter is reduced in Tfm males. HSA-AR expression increases LBM% (F) and decreases FBM% (G). Evaluation of raw values shows that HSA-AR expression has no effect on raw LBM (H), but decreases raw FBM (I). (#, Significant main effect of Tfm; *, Significant main effect of HSA-AR).

Page 175: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

165

Figure 3: No Effect of the Transgene on Body Composition in Female Rats. Transgene expression did not affect body mass (A), LBM% (B) and FBM% (C) in female rats. However, a transient effect of HSA-AR is found at 6 weeks. (*, Significant main effect of HSA-AR).

Page 176: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

166

Figure 4: Excised Fat Pads and Muscles Confirm DXA Findings. No effect of HSA-AR was found on mass of dissected individual anterior tibialis (AT – A) and extensor digitorum longus (EDL – B) muscles. However, HSA-AR male rats were found to have lighter perigonadal fat pads (C). (*, Significant difference between groups).

Page 177: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

167

Figure 5: Body Composition of HSA-AR Mice. Representative DXA images of WT and Tg L78 mice (A). Note the reduced abdominal mass in the Tg animal. L78 male mice demonstrate reduced total body mass (B), with increased LBM% (C) and decreased FBM% (D). Evaluation of raw body composition reveals decreased LBM in L78 mice (E), as well as reduced FBM (F). (*, Significant difference between groups).

Page 178: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

168

Figure 6: Effects of HSA-AR on Bone Parameters. HSA-AR expression was found to decrease BMC in Tg males (A), but not females (B). Derivation of BMD however found no difference between groups for males (C) or females (D). Similar findings were seen in HSA-AR L78 mice. Here also, transgene expression reduced BMC in Tg males (E), but had no effects on BMD (F). (*, Significant main effect of HSA-AR; #, Significant main effect of Tfm).

Page 179: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

169

Figure 7: Body Composition is Altered by T-treatment of HSA-AR Females. WT and Tg females were treated with T for 4 weeks, at which point T was removed. Body composition was measured weekly. Representative DXA images of a WT female (top) and her Tg sister (bottom) are shown over the course of treatment (A). Note the reduced abdominal mass in the Tg female at 4 weeks, and concomitant increase to baseline at 8 weeks. T treatment increases LBM% (B) and decreases FBM% (C) in Tg females only. Tg females show a T-dependent increase in raw LBM (D) and decrease in FBM (E) over the course of treatment. WT females show no differences in raw LBM (F) and FBM (G). (*Significant within-group difference from baseline for Weeks 0-4 OR Significant within-group difference from Week 4 for Weeks 5-8).

Page 180: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

170

Figure 8: Smaller Adipocytes are Found in HSA-AR Males. Representative adipocytes from WT, HSA-AR, Tfm and HSA-AR/Tfm adult males (A). Transgene expression results in reduced adipocyte size (B), with Tfm males showing the largest cells. Distribution of cell size shows that Tg animals have a larger proportion of smaller cells (C). (*, Significant difference between groups).

Page 181: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

171

Figure 9: Increased oxygen consumption in HSA-AR Male Rats. Tg adult males show increased oxygen consumption, as compared to WT controls (A). Although significant differences in RMR were not found due to HSA-AR (B), a trend toward higher levels of RMR in Tg animals was seen. (*, Significant difference between groups).

Page 182: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

172

Figure 10: Differences in Energy Expenditure in HSA-AR L78 Mice. While Tg L78 adult male mice were not found to significantly differ in oxygen consumption (A), correction for body mass reveals that L78 males have significantly higher RMR than WT brothers. (*, Significant difference between groups).

Page 183: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

173

Figure 11: HSA-AR Expression Does Not Affect Spontaneous Activity Level. Male rats were measured for activity using a laser-grid activity box, where the number of laser beam breaks was measured over a period of 1 hour. Rats were analyzed at the same ages at which differences in body composition were found. No main effect of HSA-AR or Tfm were found at any time point.

Page 184: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

174

APPENDIX A: STASTICAL VALUES

Experiment II: Body Composition Analysis MALE RATS Analysis of Variance (ANOVA) of HSA-AR Genotype (Between subjects, 2 levels) X Tfm Genotype (between subjects, 2 levels) * denotes statistical significance Main effect of HSA-AR Week 4 Body weight F=0.359 p=0.553 Lean body mass % F=1.219 p=0.278 Fat body mass % F=1.200 p=0.282 Uncorrected Lean body mass F=0.181 p=0.673 Uncorrected Fat body mass F=2.216 p=0.147 Week 6 Body weight F=0.408 p=0.528 Lean body mass % F=17.748 p<0.001* Fat body mass % F=16.275 p<0.001* Uncorrected Lean body mass F=0.154 p=0.698 Uncorrected Fat body mass F=10.541 p=0.003* Week 8 Body weight F=1.181 p=0.286 Lean body mass % F=10.630 p=0.003* Fat body mass % F=10.004 p=0.003* Uncorrected Lean body mass F=0.305 p=0.585 Uncorrected Fat body mass F=7.769 p=0.009* Week 10 Body weight F=3.339 p=0.077 Lean body mass % F=6.510 p=0.016* Fat body mass % F=6.360 p=0.017* Uncorrected Lean body mass F=0.923 p=0.344 Uncorrected Fat body mass F=6.662 p=0.015* Main effect of Tfm Week 4 Body weight F=0.817 p=0.373 Lean body mass % F=0.205 p=0.654 Fat body mass % F=0.123 p=0.728 Uncorrected Lean body mass F=0.556 p=0.461

Page 185: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

175

Uncorrected Fat body mass F=1.173 p=0.287 Week 6 Body weight F=4.517 p=0.042* Lean body mass % F=4.396 p=0.044* Fat body mass % F=4.718 p=0.038* Uncorrected Lean body mass F=3.568 p=0.068 Uncorrected Fat body mass F=7.706 p=0.009* Week 8 Body weight F=20.641 p<0.001* Lean body mass % F=27.393 p<0.001* Fat body mass % F=11.640 p=0.002* Uncorrected Lean body mass F=44.909 p<0.001* Uncorrected Fat body mass F=44.909 p<0.001* Week 10 Body weight F=22.116 p<0.001* Lean body mass % F=11.549 p=0.002* Fat body mass % F=12.837 p=0.001* Uncorrected Lean body mass F=11.560 p=0.002* Uncorrected Fat body mass F=20.914 p<0.001* Interaction between HSA-AR and Tfm Week 4 Body weight F=0.241 p=0.627 Lean body mass % F=0.207 p=0.652 Fat body mass % F=0.272 p=0.606 Uncorrected Lean body mass F=0.136 p=0.715 Uncorrected Fat body mass F=0.968 p=0.333 Week 6 Body weight F=0.448 p=0.508 Lean body mass % F=0.071 p=0.792 Fat body mass % F=0.019 p=0.891 Uncorrected Lean body mass F=0.432 p=0.516 Uncorrected Fat body mass F=0.216 p=0.645 Week 8 Body weight F=1.183 p=0.285 Lean body mass % F=0.540 p=0.468 Fat body mass % F=0.401 p=0.531 Uncorrected Lean body mass F=0.831 p=0.369 Uncorrected Fat body mass F=0.796 p=0.379 Week 10

Page 186: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

176

Body weight F=0.847 p=0.365 Lean body mass % F=0.463 p=0.501 Fat body mass % F=0.354 p=0.556 Uncorrected Lean body mass F=0.504 p=0.483 Uncorrected Fat body mass F=0.538 p=0.469 Student’s t test: Body Weight WT/WT vs WT/Tfm Wk4 p=0.994 Wk6 p=0.521 Wk8 p=0.013* Wk10 p=0.006* WT/WT vs HSA-AR/WT Wk4 p=1.000 Wk6 p=0.997 Wk8 p=0.961 Wk10 p=0.977 HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.666 Wk6 p=0.440 Wk8 p<0.001* Wk10 p<0.001* WT/Tfm vs HSA-AR/Tfm Wk4 p=0.919 Wk6 p=0.886 Wk8 p=0.765 Wk10 p=0.727 LBM% WT/WT vs WT/Tfm Wk4 p=1.000 Wk6 p=0.834 Wk8 p=0.003* Wk10 p=0.045* WT/WT vs HSA-AR/WT Wk4 p=0.992 Wk6 p=0.002* Wk8 p=0.022*

Page 187: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

177

Wk10 p=0.108 HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.850 Wk6 p=0.010* Wk8 p<0.001* Wk10 p=0.001* WT/Tfm vs HSA-AR/Tfm Wk4 p=0.799 Wk6 p<0.001* Wk8 p=0.004* Wk10 p=0.071 FBM% WT/WT vs WT/Tfm Wk4 p=1.000 Wk6 p=0.790 Wk8 p=0.001* Wk10 p=0.027* WT/WT vs HSA-AR/WT Wk4 p=0.944 Wk6 p=0.002* Wk8 p=0.024* Wk10 p=0.103 HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.865 Wk6 p=0.013* Wk8 p<0.001* Wk10 p=0.001* WT/Tfm vs HSA-AR/Tfm Wk4 p=0.781 Wk6 p<0.001* Wk8 p=0.004* Wk10 p=0.088 LBM WT/WT vs WT/Tfm Wk4 p=0.996 Wk6 p=0.775 Wk8 p=0.091 Wk10 p=0.004*

Page 188: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

178

WT/WT vs HSA-AR/WT Wk4 p=1.000 Wk6 p=0.866 Wk8 p=0.777 Wk10 p=0.164 HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.755 Wk6 p=0.115 Wk8 p=0.001* Wk10 p<0.001* HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.966 Wk6 p=0.986 Wk8 p=0.992 Wk10 p=0.108 FBM WT/WT vs WT/Tfm Wk4 p=1.000 Wk6 p=0.577 Wk8 p<0.001* Wk10 p=0.004* WT/WT vs HSA-AR/WT Wk4 p=0.961 Wk6 p=0.057 Wk8 p=0.153 Wk10 p=0.164 HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.275 Wk6 p=0.001* Wk8 p<0.001* Wk10 p<0.001* HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.482 Wk6 p=0.002* Wk8 p=0.011* Wk10 p=0.108

Page 189: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

179

FEMALE RATS Analysis of Variance (ANOVA) of HSA-AR Genotype (Between subjects, 2 levels) X Tfm Genotype (between subjects, 2 levels) * denotes statistical significance Main effect of HSA-AR Week 4 Body weight F=2.737 p=0.112 Lean body mass % F=0.485 p=0.494 Fat body mass % F=0.529 p=0.475 Week 6 Body weight F=13.992 p=0.001* Lean body mass % F=13.249 p<0.001* Fat body mass % F=14.335 p<0.001* Week 8 Body weight F=5.622 p=0.025* Lean body mass % F=.002 p= 0.968 Fat body mass % F=0.107 p=0.746 Week 10 Body weight F=2.845 p=0.163 Lean body mass % F=3.650 p=0.067 Fat body mass % F=3.044 p=0.092 Student’s t test: * denotes statistical difference LBM% WT/WT vs WT/Tfm Wk4 p= 0.025* Wk6 p=0.658 Wk8 p=0.379 Wk10 p=0.259 WT/WT vs HSA-AR/WT Wk4 p= 0.025* Wk6 p=0.004* Wk8 p=0.226 Wk10 p=.040* HSA-AR/WT vs HSA-AR/Tfm

Page 190: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

180

Wk4 p=0.141 Wk6 p=0.026* Wk8 p=0.924 Wk10 p=0.253 WT/Tfm vs HSA-AR/Tfm Wk4 p=0.653 Wk6 p=0.053 Wk8 p=0.338 Wk10 p=0.927 FBM% WT/WT vs WT/Tfm Wk4 p= 0.025* Wk6 p=0.517 Wk8 p=0.477 Wk10 p=0.203 WT/WT vs HSA-AR/WT Wk4 p=0.023* Wk6 p=0.003* Wk8 p=0.224 Wk10 p=0.039* HSA-AR/WT vs HSA-AR/Tfm Wk4 p=0.124 Wk6 p=0.0260 Wk8 p=0.9970 Wk10 p=0.238 WT/Tfm vs HSA-AR/Tfm Wk4 p=0.652 Wk6 p=0.039* Wk8 p=0.490 Wk10 p=0.821 MALE MICE Student’s t test Total body mass t=-5.594 p<0.001* LBM% t=2.94 p=0.016* FBM% t=-3.860 p=0.004* Raw LBM t=-2.944 p=0.016* Raw FBM t=-3.860 p=0.004*

Page 191: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

181

Experiment III: T-Dependence Analysis Analysis of Varience (ANOVA) of HSA-AR Genotype (Between subjects, 2 levels) X Time (within subjects, 5 levels) * denotes statistical significance ANOVAs were run separately for the 4 weeks of testosterone treatment, and the 4 weeks following cessation of treatment. For both ANOVAs, a fifth baseline week is included as a reference (Week 0 for the first and W4 for the second ANOVA). Unprotected t-tests were used only when an interaction between the within subjects factor and HSA-AR was observed. Otherwise, Dunnett’s correction was applied to alpha as indicated. ANOVA I: Weeks 0-4 LBM Within subjects LBM F=3.864 p=0.068 Between subjects HSA-AR F=7.472 p=0.015* Interaction LBM X HSA-AR F=8.863 p=0.009* Pairwise comparisons: HSA-AR W1 t=0.138 p=0.829 HSA-AR W2 t=-1.691 p=0.129 HSA-AR W3 t=-3.433 p=0.009* HSA-AR W4 t=-2.614 p=0.031* WT W1 t=-1.41 p=0.292 WT W2 t=1.251 p=0.251 WT W3 t=-0.129 p=0.901 WT W4 t=0.532 p=0.611 FBM Within subjects FBM F=991.293 p<0.001* Between subjects HSA-AR F=3.871 p=0.068 Interaction FBM X HSA-AR F=6.846 p=0.019* Pairwise comparisons: HSA-AR W1 t8=-0.133 p=0.897 HSA-AR W2 t8=1.725 p=0.123 HSA-AR W3 t8=3.406 p=0.009* HSA-AR W4 t8=2.687 p=0.028* WT W1 t7=1.242 p=0.254 WT W2 t7=-1.142 p=0.291 WT W3 t7=0.167 p=0.872 WT W4 t7=-0.455 p=0.663 Raw LBM

Page 192: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

182

Within subjects Raw LBM F=19.883 p<0.001 Between subjects HSA-AR F=0.112 p=0.742 Interaction Raw LBM X HSA-AR F=20.955 p<0.001 Pairwise comparisons: HSA-AR W1 t=-3.036 p=0.016* HSA-AR W2 t=-3.972 p=0.004* HSA-AR W3 t=-5.617 p=0.001* HSA-AR W4 t=-5.064 p=0.001* WT W1 t=-1.580 p=0.158 WT W2 t=1.126 p=0.297 WT W3 t=-0.035 p=0.973 WT W4 t=-0.534 p=0.610 Raw FBM Within subjects Raw FBM F=0.993 p=0.335 Between subjects HSA-AR F=6.977 p=0.019* Interaction Raw FBM X HSA-AR F=4.600 p=0.019* Pairwise comparisons: HSA-AR W1 t=-1.105 p=0.301 HSA-AR W2 t=0.988 p=0.352 HSA-AR W3 t=2.470 p=0.039* HSA-AR W4 t=1.625 p=0.143 WT W1 t=1.205 p=0.267 WT W2 t=-1.088 p=0.313 WT W3 t=-0.007 p=0.994 WT W4 t=-0.545 p=0.602 ANOVA II: weeks 4-8 LBM Within subjects LBM F=12.774 p=0.003* Between subjects HSA-AR F=8.826 p=0.10 Interaction LBM X HSA-AR F=0.599 p=0.451 Pairwise comparisons (Dunnett correction applied, alpha set at p<0.012): HSA-AR W5 t=0.502 p=0.629 HSA-AR W6 t=4.604 p=0.002* HSA-AR W7 t=2.337 p=0.048 HSA-AR W8 t=2.057 p=0.074 WT W5 t=-0.600 p=0.567 WT W6 t=1.369 p=0.213

Page 193: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

183

WT W7 t=1.914 p=0.097 WT W8 t=0.710 p=0.501 FBM Within subjects FBM F=13.841 p=0.002* Between subjects HSA-AR F=8.651 p=0.010* Interaction FBM X HSA-AR F=0.625 p=0.441 Pairwise comparisons (Dunnett correction applied, alpha set at p<0.012): HSA-AR W5 t=-0.484 p=0.641 HSA-AR W6 t=-4.724 p=0.001* HSA-AR W7 t=-2.350 p=0.047 HSA-AR W8 t=-2.181 p=0.061 WT W5 t=0.651 p=0.536 WT W6 t=-1.370 p=0.213 WT W7 t=1.923 p=0.096 WT W8 t=-0.740 p=0.484 Raw LBM Within subjects Raw LBM F=0.048 p=0.829 Between subjects HSA-AR F=0.002 p=0.969 Interaction Raw LBM X HSA-AR F=2.768 p=0.117 Pairwise comparisons (Dunnett correction applied, alpha set at p<0.012): HSA-AR W5 t=2.706 p=0.027 HSA-AR W6 t=5.675 p<0.001* HSA-AR W7 t=2.957 p=0.018 HSA-AR W8 t=1.670 p=0.133 WT W5 t=-0.087 p=0.933 WT W6 t=0.524 p=0.617 WT W7 t=-0.032 p=0.976 WT W8 t=-0.961 p=0.369 Raw FBM Within subjects Raw FBM F=16.980 p=0.001* Between subjects HSA-AR F=7.452 p=0.016* Interaction Raw FBM X HSA-AR F=0.50 p=0.826 Pairwise comparisons (Dunnett correction applied, alpha set at p<0.012): HSA-AR W5 t=0.143 p=0.890 HSA-AR W6 t=-3.720 p=0.006* HSA-AR W7 t=-1.872 p=0.098 HSA-AR W8 t=-1.802 p=0.109

Page 194: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

184

WT W5 t=0.324 p=0.755 WT W6 t=-1.298 p=0.236 WT W7 t=-2.036 p=0.081 WT W8 t=-1.721 p=0.129 Experiment IV: Adipose Histology Average adipocyte size Analysis of Variance (ANOVA) of HSA-AR Genotype (Between subjects, 2 levels) X Tfm Genotype (between subjects, 2 levels) * denotes statistical significance HSA-AR: F = 27.187 p<0.001* Tfm: F = 1.760 p=0.201 HSA-AR X Tfm: F = 1.282 p=0.272 Student’s t test * denotes statistical significance WT/WT vs WT/Tfm p=0.025* WT/WT vs HSA-AR/WT p=0.026* HSA-AR/WT vs HSA-AR/Tfm p=0.921 WT/Tfm vs HSA-AR/Tfm p < 0.001* Experiment V: Energy Balance and Metabolic Analyses Oxygen Consumption – Male Rats Student’s t test * denotes statistical significance WT/WT vs WT/Tfm t = 0.242; p=0.812 WT/WT vs HSA-AR/WT t = -2.025 ;p=0.056* HSA-AR/WT vs HSA-AR/Tfm t = -0.243; p=0.811 WT/Tfm vs HSA-AR/Tfm t = -2.551;p < 0.022* RMR – Male Mice Student’s t test * denotes statistical significance WT vs L78 t =-2.286; p=0.048*

Page 195: MYOCYTE ANDROGEN RECEPTOR MODULATES BODY …...Myocyte androgen receptor modulates body composition and metabolic parameters Shannon M. Fernando, Master of Science, 2010 Institute

185

Activity Box – Male Rats Analysis of Variance (ANOVA) of HSA-AR Genotype (Between subjects, 2 levels) X Tfm Genotype (between subjects, 2 levels) * denotes statistical significance 4 Weeks HSA-AR: F = 0.197; P = 0.664 Tfm: F = 0.600; P = 0.451 HSA-AR X Tfm: F = 4.277; p = 0.058 6 Weeks HSA-AR: F = 1.054; P = 0.322 Tfm: F = 2.108; P = 0.169 HSA-AR X Tfm: F = 0.575; p = 0.461 8 Weeks HSA-AR: F = 0.245; P = 0.628 Tfm: F = 2.889; P = 0.111 HSA-AR X Tfm: F = 4.114; p = 0.062 10 Weeks HSA-AR: F = 0.972; P = 0.341 Tfm: F = 1.506; P = 0.240 HSA-AR X Tfm: F = 1.603; p = 0.226