thesis cf 4

Upload: maria-ines-gregorio-martins

Post on 05-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/31/2019 thesis CF 4

    1/133

    Cystic Fibosis

    n te Gsto-Intestinl Tct

    Mjn Wotyzen-Bkke

  • 7/31/2019 thesis CF 4

    2/133

    The research described in this thesis was carried out at the Department of Pediatrics,

    Beatrix Childrens Hospital, Center for Liver, Digestive and Metabolic Diseases, University

    of Groningen, University Medical Center Groningen and was inancially supported by

    the Junior Scientiic Masterclass Groningen.

    The author gratefully acknowledges the inancial support for printing this thesis by:

    Graduate school for drug exploration University of Groningen

    University Medical Center Groningen

    ISBN 978-90-367-5090-5 (printed version)

    ISBN 978-90-367-5091-2 (digital version)

    2011 Mjn Wotyzen-Bkke

    All rights reserved. No part of this publication may be reproduced or transmitted in any

    form or by any means without permission of the author and the publisher holding the

    copyrights of the articles.

    Cover & layout design: Marjan Wouthuyzen-Bakker

    Printed by: Whrmann Print Service, Zutphen

  • 7/31/2019 thesis CF 4

    3/133

    Cystic Fibosis

    n te Gsto-Intestinl Tct

    Proefschrift

    ter verkrijging van het doctoraat in de

    Medische Wetenschappen

    aan de Rijksuniversiteit Groningen

    op gezag van deRector Magniicus, dr. E. Sterken,

    in het openbaar te verdedigen op

    woensdag 12 oktober 2011

    om 11.00 uur

    door

    Marjan Wouthuyzen-Bakker

    geboren op 7 december 1978

    te Delfzijl

  • 7/31/2019 thesis CF 4

    4/133

    Promotor: Prof. dr. H.J. Verkade

    Copromotor: Dr. H.R. de Jonge

    Beoordelingscommissie: Prof. dr. E.J. Duiverman

    Prof. dr. R.O.B. Gans

    Prof. dr. A.K. Groen

  • 7/31/2019 thesis CF 4

    5/133

    For children, the disease Cystic Fibrosis is a word that is dificult to pronounce.

    In 1965, a young boy with Cystic Fibrosis overheard his mother talking about the disease

    on the phone. When she said the words Cystic Fibrosis, he thought she said 65 roses.

    The mother was touched, because her son saw something beautiful in a disease that canoften be quite ugly. Since that time, the term 65 Roses has been used by children of all

    ages to describe their disease. The phrase is now a registered trademark of the Cystic

    Fibrosis Foundation, which adopted the rose as its symbol.

  • 7/31/2019 thesis CF 4

    6/133

    Paranimfen: Farah Klingenberg-Salachova

    Willemien de Vries

  • 7/31/2019 thesis CF 4

    7/133

    CONTENTS

    CHAPTER 1General introduction to the thesis ......................................................................................................... 7

    CHAPTER 2

    Energy intake and growth in cystic ibrosis patients with preserved pulmonary

    function .......................................................................................................................................................... 27

    Submitted

    CHAPTER 3

    Persistent fat malabsorption in cystic ibrosis; lessons from patients and mice .......... 41Journal of Cystic Fibrosis 2011; 10(3): 150-8

    CHAPTER 4

    Antibiotic treatment and fat absorption in cystic ibrosis mice ............................................ 59

    Pediatric Research, accepted for publication

    CHAPTER 5

    Ursodeoxycholic acid modulates bile low and bile salt pool independently of

    the cystic ibrosis transmembrane regulatory gene in mice ................................................... 77

    Am J Physiol Gastrointestinal Liver Physiol, conditionally accepted for publication

    CHAPTER 6

    Calcium induced anion secretion in gallbladder epithelium of pigs and mice with

    cystic ibrosis ............................................................................................................................................... 93

    Submitted

    CHAPTER 7

    General discussion and summary .....................................................................................................111

    APPENDICES

    Nederlandse samenvatting ..................................................................................................................126

    Dankwoord .................................................................................................................................................130

    Biograie .......................................................................................................................................................133

  • 7/31/2019 thesis CF 4

    8/133

  • 7/31/2019 thesis CF 4

    9/133

    ChapTEr 1

    INTRODUCTION TO THE THESIS

    M. Wouthuyzen-Bakker

  • 7/31/2019 thesis CF 4

    10/133

    10

    ChapTEr 1 INTrOduCTION

    INTrOduCTION

    Cystic Fibrosis (CF) is a complex, potentially fatal disease and affects multiple organs.

    Respiratory failure due to irreversible destruction of the lungs is the predominant cause

    of mortality in CF patients.1-2 Gastro-intestinal involvement is the second most severe

    manifestation of the disease. CF-related pancreatic and intestinal disease results in

    various problems, including malabsorption of fat and a suboptimal nutritional status.

    Biliary involvement may contribute to the former and, in addition, may also lead to end-

    stage liver disease in a subset of patients.3 The aim of this thesis was to obtain more

    detailed pathophysiological insight in CF-related gastro-intestinal disease by evaluating

    current treatment and by investigating potential future treatment options. First, the

    normal function of the CFTR protein (Cystic Fibrosis Transmembrane conductance

    Regulator) will be described. Second, the consequences of CFTR dysfunction in CF-

    disease in various organs will be outlined, with a particular emphasis on CF-related

    symptoms in the gastro-intestinal tract and corresponding treatment. In addition,

    various CF animal models will be discussed as they greatly contribute(d) to theknowledge on CF pathophysiology and related treatments.

    ThE CYSTIC FIBrOSIS TraNSMEMBraNE CONduCTaNCE rEGuLaTOr (CFTr)

    The human Cftrgene is located on the long arm of chromosome 7 and encodes for the

    Cystic Fibrosis Transmembrane conductance Regulator (CFTR). CFTR is a glycoprotein

    located at the apical membrane of (primarily) epithelial cells and is a member of the

    ATP-binding cassette (ABC) transporter family.4 CFTR is mostly expressed in gland-rich

    organs, e.g. the lungs, pancreas, intestine, gallbladder, intra- and extra-hepatic bile ducts,

    reproductive tract and sweat-glands.1-2 Recent data showed that CFTR is also localizedin non-epithelial cells and other tissues, including lymphocytes, skeletal muscle,

    smooth muscle, the thyroid gland and neurons.5-13 The most well deined function of

    CFTRcomprises its role as a chloride and bicarbonate channel. However, the knowledge

    about the function of CFTR has expanded impressively during the last decade. Apart

    from its role in anion-transport, CFTR also transports hyaluronic acid, regulates the

    activity of several other (transporter) proteins, is involved in fatty acid metabolism and

    autophagy and is (indirectly) responsible for the proper function of defensins (proteins

    with an antimicrobial function).14-20 The secretory and regulatory function of CFTR

    especially underlines its importance in luid and electrolyte transport across epithelialtissues. The CFTR protein structure comprises two transmembrane spanning domains,

    two nucleotide binding domains and one regulatory domain (Figure 1).4

    CFTR is (indirectly) activated by an increase of cyclic AMP in the cell (caused by various

    substances, including secretin). Cyclic AMP increase induces CFTR phosphorylation at

    the regulatory domain by protein kinase A.The phosphorylation stimulates the binding

    of ATP at the nucleotide binding domains and consequently results in the opening

    of the CFTR channel. Subsequently, chloride, but also bicarbonate, can be secreted

    into the lumen of the particular organ or gland. The secretion of anions generates a

    transepithelial potential difference (lumen negative) which promotes paracellular

  • 7/31/2019 thesis CF 4

    11/133

    11

    ChapTEr 1 INTrOduCTION

    sodium secretion and, consequently, osmotically driven luid secretion. Additionally,

    CFTR may act itself as a bicarbonate channel or as a chloride shunt for a separate

    chloride-bicabonate exchanger (Figure 3), resulting in active bicarbonate secretion.4

    The cascade of anion and luid secretion results in a ine water-layer at the epithelial

    surface which is responsible for the hydration of mucus and in the airways- allows the

    cilia to clear the mucus in a coordinated manner.21 Hydration of mucus and suficient

    mucus clearance prevents the harbouring of (pathogenic) bacteria and toxic/infectious

    agents to accumulate in the lumen.22 Recently, the importance of bicarbonate release

    for the expansion of mucins by electrostatic repulsion has become evident.23-26 Thus,

    bicarbonate might be essential for the formation of a normal gel-forming protective

    mucus layer. The described cascade of events after CFTR activation clearly shows the

    important role of CFTR in the protection of the epithelial lining of various glands and

    organs (Figure 3A). Inactivity or absence of CFTR, caused by various Cftrgene mutations,

    results in the disease known as: cystic ibrosis.

    Figure 1. Protein structure of the cystic ibrosis transmembrane conductance regulator(CFTR). CFTR is

    mainly located in the apical membrane of epithelial cells and consists of two transmembrane spanning domains,

    two nucleotide binding domains and one regulatory domain.

    CYSTIC FIBrOSIS (CF)

    Cystic Fibrosis (CF) is a potentially fatal, autosomal recessive disease with an incidence

    of 1:3600 life births in the Caucasian population.1-2 The prevalence of the disease

    increases due to screening of newborns and increased recognition of patients with a

    mild(er) CF phenotype.27-28 The prognosis of CF patients greatly improved during the

    last twenty years. Life expectancy increased from an average of thirty years up to forty

    years of age (Figure 2). 29 CF can be detected by a positive sweat chloride test, e.g. above

    30 mmol/L for infants below six months and above 60 mmol/L for children above

    six months of age. A second positive chloride test or the identiication of two CFTR

    mutations is conirmative for the diagnosis.30

  • 7/31/2019 thesis CF 4

    12/133

    12

    ChapTEr 1 INTrOduCTION

    CF is caused by absent, reduced or aberrant synthesis of CFTR due to Cftr gene

    mutations.31 Dysfunctional CFTR results in an impaired secretion of luid at the epithelial

    surface and consequently results in the formation of sticky and viscous mucus (which is

    the reason why CF is also referred to as mucoviscidosis).Viscous mucus obstructs and

    damages the particular organ and may allow the harbouring of pathogenic bacteria.

    The cascade of events can induce a chronic state of inlammation in various organs and

    recurrent (primarily pulmonary) infections.1-2,4

    One of the classic concepts in CF pathophysiology for the development of viscous mucus

    is the low volume hypothesis. The low volume hypothesis postulates that sodium

    hyper-absorption occurs by the loss of inhibition of the sodium channel ENaC, which

    results in the inability to hydrate the epithelial lining and results in the development of

    viscous mucus (Figure 3).32-34 This hypothesis has been recently challenged by indings in

    newborn CF pigs. In the airways of these pigs, no sodium hyper absorption or reduction

    Figure 2. Adapted from the Cystic

    Fibrosis Foundation Registry 2007

    Annual Data Report. Bethesda, Maryland.

    2009 CF foundation.

    Figure 3. Simpliied scheme

    of the function of CFTR under

    physiological and CF conditions. The

    scheme represents the classic concept

    of CF pathogenesis in bronchial

    epithelial cells. Note: (1) In gastro-

    intestinal tissues, sodium transport is

    not mediated via ENaC, but via Na+/

    H+ exchangers (NHEs) or Na+-coupled

    glucose and aminoacid transporters.

    (2) In sweat-gland ducts, CFTR serves

    to absorb chloride, not to secrete it.

    Median predicted survival age, 1985-2007

    1985 1990 1995 2000 2005

    24

    26

    28

    30

    32

    34

    36

    38

    40

    42

    year

    mediansurvivalage

    (years)

  • 7/31/2019 thesis CF 4

    13/133

    13

    ChapTEr 1 INTrOduCTION

    in the volume of the periciliary liquid layer (the water-layer at the epithelial surface)

    was present, but bacterial colonization and inability to clear bacteria was nevertheless

    observed.35-36 These indings are supported by the fact that not all tissues affected in CF

    express the epithelial sodium channel ENaC. Sodium absorption in the gastro-intestinal

    tract (e.g. small intestine, pancreas, liver and gallbladder) is not mediated via ENaC, and

    despite this, these tissues do exhibit obstruction by viscous mucus. This paradox led

    Quinton et al. to investigate the importance of bicarbonate in the pathogenesis of viscous

    mucus. 23-26 They hypothesized that bicarbonate is essential for the expansion of mucins

    and demonstrated that the absence of bicarbonate, at least under ex-vivo conditions, led

    to the formation of viscous mucus. Therefore, it is reasonable to speculate that the defect

    in bicarbonate secretion, aside chloride, is the most important factor contributing to

    CF-related disease. Unravelling the complex pathogenesis of CFTR dysfunction clearly

    remains an important challenge in CF research.

    At present, more than 1800 CFTR mutations have been identiied.37 In Europe, the deltaF508 mutation is the most common mutation found in CF patients. In the European

    population, 90% of the CF patients are heterozygous for the delta F508 mutation, while

    70% carry the mutation on both alleles.38-39 Most of the mutations can be classiied into

    one of six categories, each classiication class with a distinct functional alteration of

    CFTR (Figure 4).31

    Figure 4. Classiication of Cftr mutations. Simpliied scheme of the different classes of Cftr mutations and their

    consequences for the functionality of the CFTR protein. Class I mutations are found in ~10% of CF patients and

    include non-sense and out-frame mutations. In most of these mutations, premature truncation of normal Cftr

    translation results in the absence of synthesis of CFTR. Class II mutations include the delta F508 mutation (in-

    frame deletion), which is found in ~90% of CF patients. The deletion of phenylalanine (located in the nucleotide

    binding domain 1) causes an abnormal folding of CFTR, which is recognized by the endoplasmatic reticulum

    and is subsequently followed by degradation of the protein. Part of the misfolded/immature proteins escape

    degradation and still reaches the cell surface where residual anion secretion may be present. In class III and

    IV mutations, the CFTR reaches the cell surface, but the function of the protein is impaired by either abnormal

    gating of the ion-channel or altered conductance of ion transport respectively. In class V and VI mutations there

    is a quantitative reduction of CFTR at the cell surface. In class V mutations, reduced CFTR synthesis occurs, whilein class VI mutations, there is an increased endocytosis and degradation of CFTR in the lysosomes. 31

  • 7/31/2019 thesis CF 4

    14/133

    14

    ChapTEr 1 INTrOduCTION

    Although the severity of the disease tends to be highest in mutation class I III and

    mildest in IV - VI, an apparent genotype-phenotype association in CF patients is

    not found. Symptoms may even differ in the same family, which indicates that the

    type of mutation is not solely responsible for the CF phenotype. Modiier genes and

    environmental factors are suggested to contribute to the disease severity, especially

    concerning the pulmonary and hepato-biliary phenotype.40-43 Depending on the

    type of mutation, therapies are currently developed and tested to inhibit premature

    termination of CFTR synthesis, to correct the misfolded CFTR or to stimulate residual

    CFTR function in the apical membrane.44 The so called CFTR potentiators may restore

    gating defects and the correctors may improve CFTR protein folding and plasma

    membrane traficking. At present, VX-770, a CFTR potentiator that may be beneicial for

    patients with the CftrG551D mutation (class III), is currently tested in a phase 3 study

    with promising results.45

    CLINICaL MaNIFESTaTIONS CF aNd COrrESpONdING TrEaTMENT

    As described above, the severity and occurrence of clinical symptoms may differ among

    CF patients. We will outline the most common clinical manifestations in patients with

    classicor typical CF (diagnosed as described above) and corresponding treatment

    options. 46

    Respiratory symptoms

    Respiratory involvement is the irst cause of mortality in CF patients. 47-48 The

    accumulation of viscous and thick mucus in the distal airways results in obstruction,

    inlammation and recurrent pulmonary infections. Chronic and productive cough,dyspnoea, malaise, anorexia and weight loss often occurs, depending on the stage of

    the disease.1-2 Aggressive pulmonary intervention improved the nutritional status and

    survival of CF patients.49-53 Inhalation with bronchodilators, mucolytics and hypertonic

    saline may reduce the severity of symptoms and, inhalation with antibiotics may partly

    prevent pulmonary infections. As the disease progresses, irreversible pulmonary

    damage with a gradual decline in pulmonary functions occurs. When pulmonary

    function is severely declined (FEV1

  • 7/31/2019 thesis CF 4

    15/133

    15

    ChapTEr 1 INTrOduCTION

    of pancreatic insuficiency involves supplementation with pancreatic enzymes and

    fat soluble vitamins and a high caloric diet. Pancreatic suficient CF patients are more

    prone to the development of episodes of acute and chronic pancreatitis.56-57 In roughly

    one-third of adult patients with CF, the endocrine pancreas becomes also affected and

    results in CF-related diabetes mellitus.47

    Gastro-intestinal abnormalities

    Compared to healthy peers, symptoms of gastroesophageal relux disease are more

    described in CF patients and are mostly related to transient relaxation of the lower

    esophageal sphincter.58-59 Intestinal obstruction syndromes are a common problem in

    CF patients. Around 10-20% of CF newborns present with meconium ileus at birth, a

    condition in which the meconium (the irst stool of the infant) is thickened and can

    severely obstruct the intestine, particularly at the level of the distal small intestine.

    Meconium ileus is often the irst sign for CF. 60-61 Reversely, around 50% of infants with

    meconium ileus are diagnosed with CF.62 In older children, episodes of bowel obstruction(also known as distal ileal obstruction syndrome) and constipation may occur. Intestinal

    obstruction can be treated with oral laxatives and/or intestinal lavage, or if very severe,

    with surgical intervention. 60-61 Several secondary changes in the small intestine, like

    chronic intestinal inlammation, mucus accumulation, prolonged intestinal transit time

    and small intestinal bacterial overgrowth are also described in the CF intestine and may

    contribute to malnutrition. 63-68

    Hepatobiliary involvement

    CFTR is expressed in the gallbladder and intra- and extra-hepatic bile ducts. 69 A broadspectrum of hepato-biliary manifestations due to CFTR dysfunction has been identiied

    and may include the occurrence of gallbladder cysts, micro-gallbladder, cholelithiasis

    and cholestasis.3 Only a subset of patients (~30%) develops CF liver disease (CFLD)

    characterized by focal biliary cirrhosis, from which the course is subclinical in one-third

    of these patients.70-73 Autopsy data have indicated that, 70% of CF patients have evidence

    of focal biliary cirrhosis. Although the deinition of CFLD may vary between CF care

    centers, CFLD has been commonly deined by positive liver histology for cirrhosis or

    the presence of at least two of the following parameters: hepatomegaly, two abnormal

    serum liver enzymes or ultrasound abnormalities other than hepatomegaly (e.g.increased echogenicity, nodularity, irregular margins, splenomegaly).70 The cascade of

    decreased bile low, bile duct plugging, inlammation and focal biliary cirrhosis may

    ultimately progress to multilobar cirrhosis with portal hypertension in ~8% of CF

    patients. 71 In addition, increased bile toxicity and an altered bile salt metabolism are

    suggested to contribute to the bile duct damage.74-76 At present, end-stage CFLD accounts

    for approximately the third leading cause of death in CF patients after pulmonary

    disease and complications of lung transplantation.70 A peak incidence of CFLD occurs

    around the age of ten. It is unclear why some patients do and some patients do not

    develop CFLD. Unlike a clear genotype-phenotype relation for pancreatic involvement,

    a relation between genotype and liver disease is not so apparent. Modiier genes and

  • 7/31/2019 thesis CF 4

    16/133

    16

    ChapTEr 1 INTrOduCTION

    environmental factors are thought to play a dominant role in the development of CFLD.

    The male gender, the presence of meconium ileus at birth, pancreatic insuficiency, age

    of diagnosis and ethnic background have been identiied as risk factors for CFLD, but

    have not been conirmed in other studies. At present, only the 1-antitrypsin Z-allele

    as modiier gene has been associated with the presence CLFD, with an odds ratio of

    4.2.70,77 Ursodeoxycholic acid (UDCA) is the common medical treatment for CFLD.

    Although routinely applied in CF patients with increased levels of biochemical liver

    function tests, the therapeutic action of UDCA in CF conditions remains unclear. It has

    been hypothesized that UDCA is beneicial by its choleretic activity and/or its capacity

    to correct aberrant bile salt metabolism.78-80 No beneicial effect on survival has been

    documented, but it has been shown to improve biochemical abnormalities.81

    Other

    Since CFTR is expressed in various cell types, several other CF-related symptoms are

    described.1-2 CF patients often suffer from nasal polyps and chronic sinusitis. Especiallywhen children with CF are frequently treated with corticosteroids, osteoporosis and

    delayed puberty may occur. Male patients with CF are infertile due to obstruction of

    the vas deferens. Female patients may have problems with fertility when the cervix is

    severely obstructed with viscous mucus.

    NuTrITIONaL STaTuS

    Most children with CF have a suboptimal nutritional status. Since an impaired nutritional

    status is correlated with morbidity and mortality, optimizing the nutritional status has

    become an essential part of treatment in CF patients.82-85 The main factors contributingto the suboptimal nutritional status are malabsorption of nutrients, inadequate energy

    intake and, high energy expenditures (mainly due to recurrent pulmonary infections).

    Accordingly, cornerstones in the nutritional treatment of CF are Pancreatic Enzyme

    Replacement Therapy (PERT) for pancreas insuficiency, dietary energy intakes of

    120-150% of the Recommended Dietary Allowance (RDA) and the prevention and

    early treatment of pulmonary infections.85 Most CF infants fail to thrive at birth. After

    establishing the diagnosis of CF, supplementation with pancreatic enzymes will usually

    induce catch-up growth, but this growth spurt is often not suficient for complete

    normalization. On average, children with CF grow between the 20th

    and 30th

    percentileon the growth chart compared to healthy peers, and decline when pulmonary disease

    progresses.86-88 The implementation of neonatal screening for CF may prevent the irst

    decline in nutritional status directly after birth due to the early start of nutritional

    intervention. Because fat malabsorption persists in around 25-50% of CF patients

    despite optimal treatment with PERT,3 increased insight into the pathophysiology of CF

    related fat malabsorption is necessary to investigate new treatment strategies. Proton

    pomp inhibiters may improve the process of fat digestion and absorption in some

    individuals by reducing the duodenal acidic environment.89

  • 7/31/2019 thesis CF 4

    17/133

    17

    ChapTEr 1 INTrOduCTION

    CYSTIC FIBrOSIS aNIMaL MOdELS

    The development of several CF animal models has led to an increased understanding

    regarding the pathophysiology of CF. Until recently, CF mouse models were the only

    available animal model to study CF-related disease. CF mice proved to be very suitable

    to study intestinal disease and to evaluate novel therapeutic strategies to correct or

    potentiate defective CFTR. An important limitation of the CF mice, however, is that they

    tend to have a relatively mild CF phenotype, apart from the intestine.90-91 Most CF mouse

    models are pancreas suficient and do not exhibit CF lung or liver disease. The mild

    to absent phenotype in the organs of CF mice is in contrast to that in CF humans, and

    this has stimulated the development of other CF models in other species. Recently, the

    development of the CF pig and the CF ferret has opened a new era in CF research to

    investigate CF related pancreatic, liver and lung disease.92-95

    CF mice

    Complete Cftrknockout mice and mice with the homozygous delta F508 mutation havebeen developed by several research groups.90-91 Intestinal disease is the most prominent

    feature in CF mice. Most of the developed Cftr knockout mice have severe intestinal

    obstruction at birth, comparable to meconium ileus in the CF infant. The mortality

    rate in these mice is as high as 95%, and, dependent on the genetic background, Cftr

    knockout mice only survive on a liquid diet. The homozygous delta F508 mice still have

    residual anion secretion and therefore, exhibit a very mild intestinal phenotype. Both

    CF mouse models have impairments in the absorption of fat and increased fecal loss of

    bile salts compared to wild type littermates, but only the Cftrknockout mice also have a

    reduced body weight.96 Because CF mice are pancreas suficient, the CF mouse modelsare especially suited to gain insight into the mechanisms of pancreatic independent fat

    malabsorption.

    CF pigs

    Cftr knockout pigs and homozygous delta F508 pigs are born with meconium ileus,

    exocrine pancreas obstruction and focal biliary cirrhosis.93-95 Due to the severe intestinal

    obstruction at birth, all piglets require surgical intervention (e.g. temporarily ileostoma)

    for survival beyond neonatal age. Both CF-pig models have a micro-gallbladder, a

    condition which is also observed in approximately 30% of CF patients.3

    Although thebody weight at birth is the same as in wild type littermates, newborn Cftrknockout

    piglets fail to thrive shortly after birth, similar as seen in CF infants. No pulmonary

    abnormalities (cellular inlammation, infection or dilated or plugged submucosal

    glands) are described at birth, but characteristic features of lung disease do develop

    spontaneously with time. 93-95

    CF ferrets

    Cftr knockout ferrets display many disease characteristics similar to the pathology

    observed in CF patients, including a predisposition to pulmonary infections during the

    postnatal period, meconium ileus at birth, pancreatic insuficiency and biochemical

  • 7/31/2019 thesis CF 4

    18/133

    18

    ChapTEr 1 INTrOduCTION

    signs of liver disease.92 The biochemical parameters for liver disease (increased serum

    transaminases and bilirubin) normalize in these animals after UDCA treatment. Around

    50% of Cftr knockout ferrets with meconium ileus die after birth from intestinal

    obstruction and subsequent perforation.Additionally, even the pups without meconium

    ileus fail to thrive and die a couple of days after birth, probably because of severe

    malabsorption of nutrients. Due to the high mortality rate accompanying the very

    severe intestinal obstruction at birth, a gut corrected ferret model was developed by

    incorporating human CftrcDNA in the intestine. Although not suficient to completely

    escape the severe intestinal phenotype, one out of four of these transgenic ferrets

    passed normal stools and escaped meconium ileus. This gut corrected ferret will be

    used to expand the (recently developed) CF ferret line.92

    ThE rOLE OF aLTErNaTIVE ChLOrIdE ChaNNELS

    It has been speculated, that the marked difference in phenotypic characteristics between

    CF patients, pigs, ferrets and mice (Table 1) may be due to differences in alternative

    chloride transport.97-99 Various studies investigated the occurrence and relevance of

    several apically located alternative chloride channels in CF conditions. 97-99 Calcium

    Table 1. Organ involvement CF patients, pigs, ferrets and mice. Percentages given are estimates of the

    fraction of all CF patients/CF animals in which the pathology is manifest. The degree at which organs are affected

    may vary between animals of the same genotype. *Data on Cftr knockout mice are mainly based on observations

    in Cftr tm1Cam and Cftrtm1Unc knockout mice. Homozygous F508 mice display a very mild phenotype with

    none of the above described abnormalities, except the occurrence of diet-dependent intestinal obstruction. - : not

    (explicitly) reported in literature.

  • 7/31/2019 thesis CF 4

    19/133

    19

    ChapTEr 1 INTrOduCTION

    induced chloride channels have been considered as the most important candidate

    for alternative chloride transport in epithelial cells.100 Because the molecular identity

    of calcium induced chloride channels were unknown for quite some time, it was not

    possible to further explore and characterize their functionality in different organs. Due

    to the recent identiication of TMEM16 as a new family of chloride channels, alternative

    chloride transport regained interest in the CF ield.101-105 Alternative chloride channels

    may serve as potential rescue channel in CF conditions and could be of interest in

    therapeutic strategy to prevent or mitigate the CF phenotype.100

    SCOpE aNd OuTLINE OF ThE ThESIS

    We ultimately aim to improve the prognosis of CF by treatment of the several intestinal

    and hepato-biliary manifestations of CF disease. In this thesis we address various aspects

    of the gastro-intestinal manifestations of CF disease by evaluating current treatment

    and investigating potential future treatment options. In Cte 2, we evaluated the

    necessity of a high caloric diet in CF patients with a preserved pulmonary function.Especially during the last decades much emphasis has been placed on a high caloric

    diet in CF patients in order to improve nutritional status. We wondered whether this

    approach is necessary in all CF patients, especially those who are in a relatively mild

    state of disease. In retrospect, we cross-sectionally and longitudinally analyzed energy

    intake in this subgroup of CF patients and its relation to nutritional status and growth.

    Cte 3 provides an overview of literature on several aspects, other than pancreatic

    insuficiency, that might contribute to persistent fat malabsorption in CF patients. We

    reviewed intervention studies in CF patients and CF mice and their clinical implications.

    As antibiotic treatment has been shown to improve body weight in CF mice with smallintestinal bacterial overgrowth, we tested the hypothesis (Cte 4), that this might

    be due to improved fat absorption. We treated homozygous delta F508, Cftrknockout

    mice and wild type littermates, with broad-spectrum antibiotics and evaluated fat

    absorption. We analyzed small intestinal bacterial lora and bile salt composition,

    as these could be inluenced by antibiotic treatment and may provide mechanistic

    information on possible changes in fat absorption. In Cte 5, we addressed the

    therapeutic eficacy of ursodeoxycholate (UDCA) in CF conditions. UDCA is presently the

    only medical treatment option for CF-related liver disease, but the therapeutic action of

    UDCA in CF conditions is unclear. The postulated effect of UDCA could be mediated viaits choleretic activity or its effect on bile salt metabolism. In Cftrknockout mice and

    wild type littermates, we evaluated these options by determining several parameters

    of bile salt metabolism and bile low after UDCA treatment. Finally,we hypothesized

    that the severe gallbladder phenotype in CF pigs, in contrast to the near absence of a

    gallbladder phenotype in CF mice, is due to the differential expression and/or activity

    of alternative calcium activated chloride channels, in particular TMEM16A (Cte

    6). This hypothesis was tested experimentally by measuring trans-epithelial anion

    currents in gallbladder samples of CF mice, CF pigs and wild type littermates, following

    stimulation with CFTR-or CaCC-speciic agonists, and by monitoring expression levels

    of TMEM16A by immunocytochemistry and real-time PCR. Identifying putative rescue

  • 7/31/2019 thesis CF 4

    20/133

    20

    ChapTEr 1 INTrOduCTION

    channels in CF might lead to a therapeutic strategy to prevent or mitigate the CF

    phenotype in patients. In Cte 7 a summary of the most relevant indings will be

    discussed, including suggestions for further research.

    rEFErENCES

    1. Rowe SM, Miller S, Sorscher EJ. Cystic Fibrosis. N Eng J Med 2005;352:1992-2001.

    2. OSullivian BP, Freedman SD. Cystic Fibrosis. Lancet 2009;373:1891-904.

    3. Borowitz D, Durie PR, Lane LC et al. Gastrointestinal outcomes and confounders in cystic

    ibrosis. J Ped Gastroenterol Nutr 2005;41:273-85.

    4. Gadsby DC, Vergani P, Csanady L. The ABC protein turned chloride channel whose failure

    causes cystic ibrosis. Nature 2006;440:477-483.

    5. McDonals TV, Nghiem PT, Gardner P et al. Human lymphocytes transcribe the cystic ibrosis

    transmembrane conductance regulator gene and exhibit CF-defective cAMP-regulated

    chloride current. J Biol Chem 1992;267(5):3242-48.

    6. Lamhonwah AM, Bear CE, Huan LJ et al. Cystic ibrosis transmembrane conductance regulator

    in human muscle: dysfunction causes abnormal metabolic recovery in exercise. Ann Neurol

    2010;67(6):802-8.

    7. Li H, Ganta S, Fong P.Altered ion transport by thyroid epithelia from CFTR -/- pigs suggests

    mechanisms for hypothyroidism in cystic ibrosis. Exp Physiol 2010;95(12):1132-44.

  • 7/31/2019 thesis CF 4

    21/133

    21

    ChapTEr 1 INTrOduCTION

    8. Guo Y, Su M, McNutt MA et al. Expression and distribution of cystic ibrosis transmembrane

    conductance regulator in neurons of the human brain. J Histochem Cytochem

    2009;57(12):1113-20.

    9. Guo Y, Su M, McNutt MA. Expression and distribution of cystic ibrosis transmembrane

    conductance regulator in neurons of the spinal cord. J Neurosci Res 2009;87(16):3611-9.

    10. Su M, Guo Y, Zhao Y et al. Expression of cystic ibrosis transmembrane conductance regulator

    in paracervical ganglia. Biochem Cell Biol 2010;88(4):747-55.

    11. Pan P, Guo Y, Gu J et al. Expression of cystic ibrosis transmembrane conductance regulator in

    ganglion cells of the hearts. Neurosci Lett 2008;441(1):35-8.

    12. Mulberg AE, Weyler RT, Altschuler SM et al. Cystic ibrosis transmembrane conductance

    regulator expression in human hypothalamus. Neuroreport 1998;9(1):141-4.

    13. Michoud MC, Robert R, Hassan M et al. Role of the cystic ibrosis transmembrane conductance

    channel in human airway smooth muscle. Am J Respir Cell Mol Biol 2009;40(2):217-22.

    14. Dif F, Wu YZ, Burgel PR et al. Critical role of cytosolic phospholipase A2 in bronchial mucushypersecretion in CFTR-deicient mice. Eur Respir J 2010;36(5):1120-30.

    15. Luciani A. Villella VR, Esposito S et al. Defective CFTR induces aggresome formation and lung

    inlammation in cystic ibrosis through ROS-mediated autophagy inhibition. Nat Cell Biol

    2010;12(9):863-75.

    16. Strandvik B. Fatty acid metabolism in cystic ibrosis. 2010;83(3):121-9.

    17. Schulz T, Schumacher U, Prante C et al. Cystic ibrosis transmembrane conductance regulator

    can export hyaluronan. Pathobiology 2010;77(4):200-9.

    18. Ollero M, Janaidi O, Zaman MM et al. Decreased expression of peroxisome proliferator

    activated receptor gamma in Cftr -/- mice. J Cell Physiol 2004;200(2):235-44.

    19. Harmon GS, Dumlao DS, Ng DT et al. Pharmacological correction of a defect in PPAR-gamma

    signaling ameliorates disease severity in Cftr-deicient mice. Nat Med 2010;16(3):313-8.

    20. Goldman MJ, Anderson GM, Stolzenberg ED et al. Human beta-defensin-1 is a salt-sensitive

    antibiotic in lung that is inactivated in cystic ibrosis. Cell 1997;88(4):553-60.

    21. Matsui H, Randell SH, Peretti SW et al. Coordinated clearance of peiciliary liquid and mucus

    from airway surfaces. J Clin Invest 1998;102(6):1125-31.

    22. Houtmeyers E, Gosselink R, Gayan-Ramirez G et al. Regulation of mucociliary clearance in

    health and disease. Eur Resp J 1999;13(5):1177-88.

    23. Garcia MA, Yang N and Quinton PM. Normal mouse intestinal mucus release requires

    cystic ibrosis transmembrane regulator-dependent bicarbonate secretion. J Clin Invest

    2009;119(9):2613-22.

    24. Quinton PM. Role of epithelial HCO3- transport in mucin secretion: lessons from cystic ibrosis.

    Am J Physiol Cell Physiol 2010;299(6):C1222-33.

    25. Quinton PM. Birth of mucus. Am J Physiol Lung Cell Mol Physiol 2010;298(1):L13-4.

    26. Quinton PM. Cystic ibrosis: impaired bicarbonate secretion and mucoviscidosis. Lancet

    2008;372:415-17.

    27. Rock MJ. Newborn screening for cystic ibrosis. Clin Chest Med 2007;28:297-305.

  • 7/31/2019 thesis CF 4

    22/133

    22

    ChapTEr 1 INTrOduCTION

    28. Strausbaugh SD, Davis PB. Cystic ibrosis: a review of epidemiology and pathobiology. Clin

    Chest Med 2007;28:279-88.

    29. Cystic Fibrosis Foundation. Patient Registry Annual Report2007. Bethesda, MD: Cystic

    Fibrosis Foundation; 2007.

    30. Farrell PM, Rosenstein BJ, White TB et al. Guidelines for diagnosis of cystic ibrosis in newbornsthrough older adults: Cystic Fibrosis Foundation Consensus report. J Pediatr 2008;153:S414.

    31. Rowntree RK, Harris A. The phenotypic consequences of CFTR mutations. Ann Hum Genet

    2003;67:471-485.

    32. Matsui H, Grubb BR, Tarran R et al. Evidence for periciliary liquid layer depletion, not abnormal

    ion composition, in the pathogenesis of cystic ibrosis airway disease. Cell 1998;95:1005-15.

    33. Matsui H, Wagner VE, Hill DB et al.A physical linkage between cystic ibrosis airway surface

    dehydration and Pseudomonas aeruginosa bioims. Proc Natl Acad Sci USA 2006;103:18131-

    36.

    34. Boucher RC.Airway surface dehydration in cystic ibrosis: pathogenesis and therapy. .AnnuRev Med 2007;58:157-70.

    35. Chen JH, Stoltz DA, Karp PH et al. Loss of anion transport without increased sodium absorption

    characterizes newborn porcine cystic ibrosis airway epithelia. Cell 2010;143(6):911-23.

    36. Cystic ibrosis: an ion transport issue?Nat Med 2011;17(2):166-67.

    37. Cystic ibrosis genetic analysis consortium. Cystic ibrosis mutation database. Available at:

    http://www.genet.sickkids.on.ca /cftr/StatisticsPage.html.

    38. Estivill X, Bancells C, Ramos C. Geographic distribution and regional origin of 272 cystic

    ibrosis mutations in European populations. Human Mutat 1997;10:135-54.

    39. Farrell PM. The prevalence of cystic ibrosis in the European Union. J Cyst Fibros. 2008:7:450-

    53.

    40. Bartlett JR, Friedman KJ, Ling SC et al. Genetic modiiers of liver disease in cystic ibrosis.

    JAMA 2009;302(10):1076-83.

    41. Merlo CA, Boyle MP. Modiier genes in cystic ibrosis lung disease. J Lab Clin Med

    2003;141(4):237-41.

    42. Cutting GR. Modiier genes in Mendelian disorders: the example of cystic ibrosis . Ann N Y

    Acad Sci 2010;1214:57-69.

    43. Collaco JM, Cutting GR. Update on gene modiiers in cystic ibrosis. Curr Opin Pulm Med

    2008;14(6):559-66.

    44. Sloane PA, Rowe SM. Cystic ibrosis transmembrane conductance regulator protein repair as

    a therapeutic strategy in cystic ibrosis. Curr Opin Pulm Med 2010;16:591-97.

    45. Accurso FJ, Rowe SM, Clancy JP et al. Effect of VX-770 in persons with cystic ibrosis and the

    G551D-CFTR mutation. N Engl J Med 2010;363(21):1991-2003.

    46. De Boeck K, Wilschanski M, Catellani C et al. Cystic ibrosis: terminology and diagnostic

    algorithms. Thorax 2006;61:627-35.

    47. Anon. 2006 Annual data report to the center directors. Bethesda, MD: Cystic ibrosis

    foundation patient registry, 2007.

  • 7/31/2019 thesis CF 4

    23/133

    23

    ChapTEr 1 INTrOduCTION

    48. Dodge JA, Lewis PA, Stanton M et al. Cystic ibrosis mortality and survival in the UK: 1947-

    2003. Eur Respir J 2007; 29:522-26.

    49. Cohen-Cymberknoh M, Shosesov D, Kerem E. Managing cystic ibrosis: strategies that

    increase life expectancy and improve quality of life. Am J Respir Crit Care Med 2011; in press.

    50. Padman R, McColley SA, Miller DP et al. Infant care patterns at epidemiologic study of cysticibrosis sites that achieve superior childhood lung function. Pediatrics 2007;119:E531-37.

    51. Johnson C, Butler SM, Konstan MW et al. Factors inluencing outcomes in cystic ibrosis: a

    center-based analysis. Chest 2003;123:20-27.

    52. Szaff M, Hoiby N, Flensborg EW. Frequent antibiotic therapy improves survival of cystic

    ibrosis patients with chronic Pseudomonas Aeruginosa infection. Acta Paediatr Scand

    1983;72:651-57.

    53. Regelmann WE, Elliot GR, Warwick WJ et al. Reduction of sputum Pseudomonas Aeruginosa

    density by antibiotics improves lung function in cystic ibrosis more than do bronchodilators

    and chest physiotherapy alone. Am Rev Respir Dis 1990;141:914-21.

    54. Aurora P, Spencer H, Moreno-Galdo. Lung transplantation in children with cystic ibrosis: a

    view from Europe. Am J Respir Crit Care Med 2008;177:935-36.

    55. Couper RT, Corey M, Moore DJ et al. Decline of exocrine pancreatic function in cystic ibrosis

    patients with pancreatic suficiency. Pediatr Res 1992;32:179-82.

    56. Augarten, A, Ben Tov, A, Madgar, I, et al. The changing face of the exocrine pancreas in cystic

    ibrosis: the correlation between pancreatic status, pancreatitis and cystic ibrosis genotype.

    Eur J Gastroenterol Hepatol 2008; 20:164.

    57. De Boeck, K, Weren, M, Proesmans, M, et al. Pancreatitis among patients with cystic ibrosis:

    correlation with pancreatic status and genotype. Pediatrics 2005; 115:e463.

    58. Malfroot A, Dab I. New insights on gastro-oesophageal relux in cystic ibrosis by longitudinal

    follow up. Arch Dis Child. 1991;66(11):1339-45.

    59. van der Doef HP, Arets HG, Froeling SP et al. Gastric acid inhibition for fat malabsorption or

    gastroesophageal relux disease in cystic ibrosis: longitudinal effect on bacterial colonization

    and pulmonary function. J Pediatr. 2009;155(5):629-33.

    60. Houwen RH, van der Doef HP, Sermet I et al. Intestinal obstruction syndromes in cystic ibrosis:

    meconium ileus, distal intestinal obstruction syndrome and constipation. Curr Gastroenterol

    Rep 2011, in press.

    61. Raia V, Fuiano L, Staiano A. Deining DIOS and constipation in cystic ibrosis with a multicentre

    study on the incidence, characteristics and treatment of DIOS. J Pediatr Gastroenterol Nutr.

    2010;50(1):38-42.

    62. Gorter RR, Karimi A, Sleeboom C. Clinical and genetic characteristics of meconium ileus in

    newborns with and without cystic ibrosis. J Ped Gastroenterol Nutr 2010;50(5):569-72.

    63. Lewindon PJ, Robb TA, Moore DJ et al. Bowel dysfunction in cystic ibrosis: importance of

    breath testing. Journal of Paediatrics and Child Health 1998;34(1):79-82.

    64. Lisowska A, Wjtowicz J, Walkowiak J. Small intestine bacterial overgrowth is frequent in

    cystic ibrosis: combined hydrogen and methane measurements are required for its detection.

    Acta Biochimica Polonica 2009;56(4):631-34.

  • 7/31/2019 thesis CF 4

    24/133

    24

    ChapTEr 1 INTrOduCTION

    65. Bruzzese ER, Raia V, Gaudiello G et al. Intestinal inlammation is a frequent feature of

    cystic ibrosis and is reduced by probiotic administration. Alimentary Pharmacology and

    Therapeutics 2004;20(7):813-19.

    66. Raia V, Maiuri L, de Ritis G et al. Evidence of chronic inlammation in morphologically normal

    small intestine of cystic ibrosis patients. Pediatric Research 2000;47(3):344-50.

    67. Sbarbati A, Bertini M, Catassi C et al. Ultrastructural lesions in the small bowel of patients

    with cystic ibrosis. Pediatric Research 1998;43(2):234-39.

    68. Smyth RL, Croft NM, OHea U et al. Intestinal inlammation in cystic ibrosis. Arch of Dis in

    Child 2000;82(5):394-99.

    69. Cohn JA, Strong TV, Picciotto MR, et al. Localization of the cystic ibrosis transmembrane

    conductance regulator in human bile duct epithelial cells. Gastroenterology 1993;105:1857-

    1864.

    70. Colombo C, Battezzati PM, Crosignani A, et al. Liver disease in cystic ibrosis: A prospective

    study on incidence, risk factors, and outcome. Hepatology 2002;36:1374-1382.

    71. Nash KL, Allison ME, McKeon D, et al.A single centre experience of liver disease in adults with

    cystic ibrosis 1995-2006. J.Cyst.Fibros. 2008;7:252-257.

    72. Herrmann U, Dockter G, Lammert F. Cystic ibrosis-associated liver disease. Best Pract Res

    Clin Gastroenterol 2010;24:585-92.

    73. Colombo C, Battezzati PM. Liver involvement in cystic ibrosis: primary organ damage or

    innocent bystander?J Hepatol 2004;41:1041-44.

    74. Lindblad A, Hultcrantz R, Strandvik B. Bile-duct destruction and collagen deposition: a

    prominent ultrastructural feature of the liver in cystic ibrosis. Hepatology 1992;16:372-81.

    75. Freudenberg F, Broderick AL, Yu BB et al. Pathophysiological basis of liver disease incystic ibrosis employing a deltaF508 mouse model. J Physiol Gastrointest Liver Physiol

    2008;294:G1411-20.

    76. Smith JL, Lewindon PJ, Hoskins AC et al. Endogenous ursodeoxycholic acid and cholic acid in

    liver disease due to cystic ibrosis. Hepatology 2004;39:1673-82.

    77. Wilschanski M, Rivlin J, Cohen S et al. Clinical and genetic risk factors for cystic ibrosis-

    related liver disease. Pediatrics 1999;103:52-7.

    78. Fiorotto R, Spirli C, Fabris L, et al. Ursodeoxycholic acid stimulates cholangiocyte luid

    secretion in mice via CFTR-dependent ATP secretion. Gastroenterology 2007;133:1603-1613.

    79. Shimokura GH, McGill JM, Schlenker T, et al. Ursodeoxycholate increases cytosolic calciumconcentration and activates Cl- currents in a biliary cell line. Gastroenterology 1995;109:965-

    972.

    80. Gurantz D,.Hofmann AF. Inluence of bile acid structure on bile low and biliary lipid secretion

    in the hamster. Am.J.Physiol 1984;247:G736-G748.

    81. Cheng K, Ashby D, Smyth R. Ursodeoxycholic acid for cystic ibrosis-related liver disease.

    Cochrane Database Syst Rev 2000;(2):CD000222.

    82. Sinaasappel M, Stern M, Littlewood J et al. Nutrition in patients with cystic ibrosis: a

    European Consensus. J Cyst Fibros 2002;1(2):51-75.

  • 7/31/2019 thesis CF 4

    25/133

    25

    ChapTEr 1 INTrOduCTION

    83. Beker LT, Russek-Cohen E, Fink RJ. Stature as a prognostic factor in cystic ibrosis survival. J

    Am Diet Assoc 2001;101(4):438-442.

    84. Corey M, McLaughlin FJ, Williams M, et al.A comparison of survival, growth, and pulmonary

    function in patients with cystic ibrosis in Boston and Toronto. J Clin Epidemiol 1988;

    41(6):583-591

    85. Sinaasappel M, Stern M, Littlewood J, et al. Nutrition in patients with cystic ibrosis: a

    European Consensus. J Cyst Fibros 2002;1(2):51-75.

    86. Haeusler G, Frisch H, Waldhor T et al. Perspectives of longitudinal growth in cystic ibrosis

    from birth to adult age. Eur J Pediatr 1994;153(3):158-163.

    87. Patel L, Dixon M, David TJ. Growth and growth charts in cystic ibrosis. J R Soc Med 2003;96

    Suppl 43:35-41.

    88. Stettler N, Kawchak DA, Boyle LL et al. Prospective evaluation of growth, nutritional status,

    and body composition in children with cystic ibrosis. Am J Clin Nutr 2000;72(2):407-413.

    89. Jones A. Drug therapies for reducing gastric acidity in people with cystic ibrosis . TheCochrane collaboration 2003;2:1-16.

    90. Guibalt C, Saeed Z, Downey GP, Radzioch D. Cystic ibrosis mouse models. Am J Respir Cell

    Mol Biol 2007;36:1-7.

    91. Scholte BJ, Davidson DJ, Wilke M, De Jonge HR.Animal models of cystic ibrosis. J Cyst Fibr

    2004;3:183-190.

    92. Sun X, Sui H, Fisher JT et al. Disease phenotype of a ferret CFTR-knockout model of cystic

    ibrosis. J Clin Invest 2010;120(9):3149-60.

    93. Meyerholz DK, Stoltz DA, Pezzulo AA, Welsh MJ. Pathology of gastro-intestinal organs in a

    porcine model of cystic ibrosis. Am J Path 2010;176(3):1-13.

    94. Rogers CS, Stoltz DA, Meyerholz DK et al. Disruption of the CFTR gene produces a model of

    cystic ibrosis in newborn pigs. Science 2008;321:1837-40.

    95. Ostedgaard LS, Meyerholz DK, Chen JH et al. The F508 mutation causes CFTR misprocessing

    and cystic ibrosis-like disease in pigs. Sci Transl Med 2011; in press.

    96. Bijvelds MJ, Bronsveld I, Havinga R, et al. Fat absorption in cystic ibrosis mice is impeded

    by defective lipolysis and post-lipolytic events. Am J Physiol Gastrointest Liver Physiol

    2005;288:G646-G653.

    97. Zdebik AA, Cuffe JE, Bertog M et al. Additional disruption of the ClC-2 Cl- channel does

    not exacerbate the cystic ibrosis phenotype of cystic ibrosis transmembrane conductanceregulator mouse models. J Biol Chem 2004;279(21):22276-83.

    98. Bijvelds MJ, Bot AG, Escher JC et al.Activation of intestinal Cl- secretion by lubiprostone requires

    the cystic ibrosis transmembrane conductance regulator. Gastroenterol 2009;137(3):976-

    85.

    99. Jentsch TJ. CLC chloride channels and transporters: from genes to protein structure, pathology

    and physiology. Crit Rev Biochem Mol Biol 2008;43(1):3-36.

    100. Verkman AS, Galietta LJV. Chloride channels as drug targets. Nat Rev 2009;8:153-68.

    101. Yang YD, Cho H, Koo JY et al. TMEM16A confers receptor-activated calcium-dependent

    chloride conductance. Nature 2008;435:1210-1216.

  • 7/31/2019 thesis CF 4

    26/133

    26

    ChapTEr 1 INTrOduCTION

    102. Schroeder BC, Cheng T, Jan YN et al. Expression cloning of TMEM16A as a calcium-activated

    chloride subunit. Cell 2008;134:1019-1029.

    103. Huang F, Rock JR, Harfe BD, et al. Studies on expression and function of the TMEM16A calcium-

    activated chloride channel. Proc Natl Acad Sci U.S.A. 2009;106:21413-18.

    104. Galietta LJV. The TMEM16A protein family: a new class of chloride channels. Biophys J2009;97:3047-3053.

    105. Caputo A, Caci E, Ferrera L, et al. TMEM16A, a membrane protein associated with calcium

    dependent chloride channel activity. Science 2008;322:590-593.

  • 7/31/2019 thesis CF 4

    27/133

    ChapTEr 2

    ENERGY INTAKE AND GROWTH

    IN CYSTIC FIBROSIS PATIENTS WITH

    PRESERVED PULMONARY FUNCTION

    M. Wouthuyzen-Bakker1

    C.K. van der Ent2

    J.W. Woestenenk3

    M. Zwolsman1

    H.J. Verkade1

    F.A.J.A. Bodewes1

    1 Pediatric Gastroenterology, Department of Pediatrics, University Medical Center

    Groningen, University of Groningen, Groningen, The Netherlands.2 Department of Pediatrics, University Medical Center Utrecht, Utrecht,

    the Netherlands3 Julius Centre for Health Sciences and Primary Care, Department of Dietetics and

    Nutritional Science, University Medical Center Utrecht, Utrecht, the Netherlands

    Sbmitte

  • 7/31/2019 thesis CF 4

    28/133

    28

    ChapTEr 2 INTaKE aNd GrOWTh

    aBSTraCT

    Improving the nutritional status remains an important challenge in pediatric cystic

    ibrosis (CF) patients. The effect of a high energy intake on the nutritional status in

    relatively healthy pediatric CF patients is not well described. We investigated the

    relation between energy intake, nutritional status and growth in school-age CF patients

    with preserved pulmonary function. In a cohort of Dutch CF patients, we analyzed

    data of 81 children with FEV1-values >80%. Patients below 6 and above 9 years were

    excluded to avoid inaccurate spirometric outcomes and growth interference during

    puberty respectively. Wecross-sectionally assessed the relation between energy intake,

    nutritional status and growth (multiple-regression) and longitudinally evaluated the

    effect of changes in energy intake on weight (paired t-test). The nutritional status

    was slightly below average compared to the healthy reference population (z-score

    weight:-0.61.8, BMI:-0.10.7). Upon cross-sectional analysis, we found no positive

    relation between energy intake and nutritional status or growth. Upon longitudinal

    analysis, 19 patients increased their energy intake (10.29.8 kcal/kg/bw) in the yearafter the initial assessment, but the z-score for weight improved (0.180.16) in only

    20% of the 19 patients. The positive effect of energy intake on weight was irrespective

    from the initial z-score for weight or the initial energy intake. Conclusion. The relation

    between energy intake, nutritional status and growth is weak in school-age CF patients

    with preserved pulmonary function. The results emphasize the need for individualized

    nutritional treatment, but simultaneously underline the dificulty to design nutritional

    trials with suficient statistical power in this subgroup of CF patients.

  • 7/31/2019 thesis CF 4

    29/133

    29

    ChapTEr 2 INTaKE aNd GrOWTh

    INTrOduCTION

    Cystic Fibrosis (CF) is an autosomal recessive disease and is caused by a mutation on

    the Cftrgene located on chromosome 7, which encodes for a chloride and bicarbonate

    channel in, particularly, epithelial cells. A defective CFTR protein results in the

    formation of viscous and sticky mucus with bacterial colonization, inlammation and

    eventually damage to different glands and gland rich organs. Intestinal malabsorption

    of nutrients (mainly due to pancreatic insuficiency), inadequate energy intake and

    increased energy expenditure due to recurrent pulmonary infections all contribute

    to a suboptimal nutritional status in CF patients. 28 Since nutritional status is related

    to morbidity and mortality in CF, improving the nutritional status remains one of the

    important challenges in CF care.

    The well known epidemiological study in Boston and Toronto of Corey et. al. in the late

    80s, introduced a major shift in the nutritional treatment of CF. The formerly restricted

    fat diet adviced for CF patients, was changed into a high fat diet. Another major changecomprised more aggressive pulmonary treatment and prevention of weight loss

    during infections. This renewed approach resulted in an improved nutritional status

    and overall survival of CF patients.2, 6, 8, 26 Consequently, the current general CF dietetic

    recommendations comprises high dietary energy intakes ranging from 110-200% of

    the Recommended Daily Allowance (RDA) in patients with pancreatic insuficiency, in

    which energy derived from fat should be approximately 40%.28,29

    However, many children with CF have dificulties to meet the dietary recommendations.

    24,25 This phenomenon often creates a burden for CF children, parents and health careproviders in their attempt to comply to the general recommendation. Nowadays,

    several studies report on adverse social and psychological consequences for CF patients

    and their families aiming to achieve high energy intakes, like family stress and feeding

    dificulties.7, 16, 23, 39

    Since there are no conclusive studies which exclusively substantiated the potential

    beneicial effects of a high dietary energy intake in CF patients with preserved

    pulmonary function, we aimed to investigate the relation between dietary energy intake

    and nutritional status and growth in this subgroup of patients.

    paTIENTS aNd METhOdS

    We retrospectively evaluated data of pediatric CF patients who were under medical

    control on the 1st of January 2008 in the University Medical Centers in Utrecht and

    Groningen in the Netherlands. Both centers are specialized regional CF care centers.

    Patients visited the CF Center, at least annually, for a complete check up, consisting of a

    72 hours fecal fat balance test, lung function test and a sputum culture test (if obtained).

    Children with FEV1 values above 80% were included in the study. Patients below

    6 and above 9 were excluded to avoid inaccurate spirometry outcomes.10, 20, 37 and to

  • 7/31/2019 thesis CF 4

    30/133

    30

    ChapTEr 2 INTaKE aNd GrOWTh

    avoid pubertal growth effects respectively. For our cross-sectional analysis (n=81), the

    irst recorded measurement for each child was used. In order to evaluate the effect of

    energy intake on weight (longitudinally), children from which complete datasets of at

    least two time points were available were included in the study (n=44) (Figure 1). The

    excluded children (n=37) had similar patient characteristics (parameters nutritional

    status, mutation class, co-morbidity etc) as the included children (data not shown).

    This study was deemed exempt by the University Medical Center Groningen and Utrecht

    Institutional Review Board.

    Assessment of nutritional status

    Weight (kg), length (cm) and BMI measurements of every child were collected. Annual

    growth velocity for length and weight were calculated using the charts of Tanner and

    Whitehouse. 35 Growth velocity was deined as the increment in the childs length (cm/

    yr) and weight (kg/yr) divided by the time interval between the two measurements(with a minimum of 9 months and a maximum of 18 months). All growth parameters

    were converted into age-corrected z-scores based on Dutch national growth studies

    using Growth analyzer 3 software of the Dutch Growth Foundation.9

    Assessment of energy intake

    A three-day dietary diary was used to determine dietary intake of total energy. The

    dietary diaries contained three day prospective recorded household measurements

    with detailed information on macronutrient intake. We assumed that the dietary diary

    relected the average daily dietary energy intake in the preceding year. All dietary

    diaries were manually processed by experienced specialized CF dieticians. The dietary

    Figure 1. Flow chart.

    Flow chart for the retrospective

    analysis in pediatric CF patients

    between the age of 6-9 years with

    preserved pulmonary function

    (FEV1>80%). For the cross-sectional

    analysis, the irst measurement of

    each child was used (n=81). For

    the longitudinal analysis, data

    available with at least two time

    points per child was used. The irst

    two measurements per time pointwere selected (n=44). FEV1: Forced

    Expiratory Volume in 1 second.

    dataset 1999-2008

    657 measurements

    n = 268

    FEV1 > 80%

    440 measurements

    n = 183

    FEV1 < 80%

    217 measurements

    n = 108

    6-9 years

    43 measurements

    n = 31

    6-9 years

    150 measurements

    n = 81

    2 measurements per child:

    n = 44

  • 7/31/2019 thesis CF 4

    31/133

  • 7/31/2019 thesis CF 4

    32/133

    32

    ChapTEr 2 INTaKE aNd GrOWTh

    Statistical analysis

    Statistical analysis was performed using SPSS 16.0 (SPSS Inc., Chicago, IL).Cross-sectional

    analysis. The relation between energy intake and nutritional status and growth was

    cross-sectionally assessed by linear regression analysis. A stepwise multiple regression

    analysis was performed to correct for confounding effects on nutritional status and

    growth. The analysis included z-scores for weight, length, BMI, weight gain and length

    growth as dependent variablesand were tested for normal distribution.Total energy

    intake, fat absorption (%), mutation class, age and gender were used as independent

    variables. We repeated the analysis and replaced the variable total energy intake for the

    variable energy intake of fat. The differentCftrmutation classes were included in the

    multiple regression analysis by transforming them into dummies and by using the enter

    method. Longitudinal analysis. We longitudinally evaluated the effect of an increased or

    decreased energy intake (compared to the previous year) on the z-score for weight by

    using the paired student t-test. All data are reported as means the standard deviation

    (SD). P-values < 0.05 were considered as statistically signiicant.

    rESuLTS

    Characteristics study group

    Table 1 shows the patient characteristics for the cross-sectional as the longitudinal

    analysis for several parameters. Parameters were similar between the cross-sectional

    and the longitudinal data. All patients within our cohort were pancreas insuficient and

    were treated with pancreatic enzymes. The overall nutritional status was slightly belowaverage compared to the healthy reference population according to length, weight and

    BMI. Growth velocity scores were above average for length as well as weight. The mean

    total energy intake and the percentual energy intake derived from fat in our cohort of

    pediatric CF patients was comparable to the general CF nutritional recommendations

    of 120-150% [28]. Except for meconium ileus, with a higher incidence in males, gender

    was equally distributed among the other parameters.

    Relation between energy intake and nutritional status (cross-sectional analysis)

    Although the mean z-scores for nutritional status in our cohort of patients was slightlybelow average compared to the healthy reference population (Table 1), the z-score for

    weight was below zero in 78% of patients (Figure 3). We found no relation between

    total energy intake or fat intake and z-scores for weight, length, BMI, weight gain and

    length growth (Figure 3). A weak negative association was found between total energy

    intake and the z-score for weight (R=0.27, p=0.02). The multiple regression analysis,

    that included several potential confounding factors on nutritional status or growth (fat

    absorption, mutation class, age and gender), showed no dependency on these results.

    The z-score for BMI was positive associated with age (R= 0.22, p=0.03).

  • 7/31/2019 thesis CF 4

    33/133

    33

    ChapTEr 2 INTaKE aNd GrOWTh

    Table 1. Patient characteristics. Baseline characteristics of cross-sectional and longitudinal data of pediatric

    cystic ibrosis patients with preserved pulmonary function (deined as FEV1 values > 80%). Z-scores for weight

    and length are depicted as z-scores for age. Cross-sectional and longitudinal values for all parameters were

    not statistically different from each other. Ursodeoxy-cholate was prescribed because of suspicion of CF related

    liver disease, based on persistent increased serum trans-aminases, hepato-megaly and/or hepatic ultrasound

    abnormalities (40-41). CFTR gene mutations were divided in ive classes based on their demonstrated orpresumed molecular consequences (19).

    FEV1: Forced Expiratory Volume in 1 second. RDA: Recommended Daily Allowance. Kcal/kg/bw: kilocalories per

    kilogram body weight. Parameters are reported as a percentage or as mean SD.

  • 7/31/2019 thesis CF 4

    34/133

    34

    ChapTEr 2 INTaKE aNd GrOWTh

    The effect of an increased or decreased energy intake on z-score weight (longitudinal

    analysis)

    In correspondence with the cross-sectional results, we found no relation between energy

    intake and the z-score for weight in the longitudinal analysis (Figure 4). Out of the 44

    patients, 17 increased their energy intake in the year after the initial assessment (mean

    increase 10.29.8 kcal/kg/bw), but only 4 of these patients showed a corresponding

    improvement in the z-score for weight (right upper quadrant in Figure 4A), while

    the majority of patients showed a stabilization or decrease in the z-score for weight

    (right lower quadrant in Figure 4A). A decrease in energy intake in the year after the

    initial assessment (mean decrease 13.39.5 kcal/kg/bw) in 27 of patients, resulted in a

    corresponding decrease in the z-score for weight in roughly 50% of these patients, while

    the other 50% of patients showed an improvement (left lower and upper quadrant inFigure 4A, respectively).

    Figure 3. Relation between energy intake and nutritional status (cross-sectional analysis).

    Upper panel: relation between total energy intake and z-scores for weight (A), BMI (B), and weight gain (C).

    Lower panel: relation between fat intake and z-scores for weight (D), BMI (E), and weight gain (F). kcal/kg/bw:

    kilocalories per kilogram body weight. R: correlation coeficient.

  • 7/31/2019 thesis CF 4

    35/133

    35

    ChapTEr 2 INTaKE aNd GrOWTh

    To examine these results in more detail, we additionally investigated whether the

    effect of an increased or decreased energy intake on weight after a year of the initial

    assessment, depended on the initial z-score for weight or on the initial amount of

    energy intake. We subdivided patients with initial z-scores for weight above or below

    0. The subdivision did not affect the longitudinal relation between energy intake and

    z-score for weight (Figure 5). We found a decrease in z-score for weight in patients

    with initial high z-scores for weight during increased energy intake (n=5; Figure 5B).

    When subdividing patients with initial energy intakes above and below 90 kilocalories

    per kilogram body weight, we neither found a difference between an increased or a

    decreased energy intake concerning the effect on the z-score for weight (data not

    shown).

    Figure 4. Relation between delta energy intake and delta z-score for weight (longitudinal analysis).

    A) Relation between the difference () in annual energy intake and the corresponding annual difference in z-score

    for weight. B) Percent distribution of patients with increased or decreased energy intake after a year of the initial

    assessment and the corresponding effect on the z-score for weight. kc/kg/bw: kilocalories per kilogram body

    weight. R: correlation coeficient. : increased, : decreased, : not changed.

    Figure 5. The effect of an increased or

    decreased energy intake on z-score

    weight.

    In order to conclude about the effect of

    an increased (A-B) or decreased (C-D)

    energy intake in relation to the initial

    nutritional status of CF patients, patients

    were categorized according to their initial

    z-score of weight (below or above 0) at

    time point 0 (T0). Figures A and C depicts

    the actual increase or decrease in energy

    intake in kilocalories per kilogram body

    weight (kcal/kg/bw) in the following year

    (T1) . Figures B and D depicts the effect of

    the increased or decreased energy intake

    on z-score weight in the following year

    (T1). Values are depicted as mean SD. *: p-value < 0.05 (comparing T0 with T1).

  • 7/31/2019 thesis CF 4

    36/133

    36

    ChapTEr 2 INTaKE aNd GrOWTh

    dISCuSSION

    Our study is the irst which exclusively investigated the relation between energy intake

    and nutritional status and growth in school-age pediatric CF patients with a preserved

    pulmonary function. We found that approximately 80% of CF children have a z-score for

    weight below zero, despite preservation of pulmonary function. However, retrospective

    evaluation, in a cross-sectional and longitudinal setting, showed no relation between

    energy intake and nutritional status or growth.

    Until now, most studies evaluated the relation between energy intake and either

    nutritional status or growth in pediatric CF patients who already are in a more advanced

    stage of disease 1, 4, 13, 15, 18-19, 22, 27, 30-33, 36, 38 (e.g. patients with impaired pulmonary function

    and/or patients who are more severely malnourished). From retrospective studies we

    learned that, in this group of CF patients, a weak correlation (R

  • 7/31/2019 thesis CF 4

    37/133

    37

    ChapTEr 2 INTaKE aNd GrOWTh

    subgroup of patients; all of our patients were pre-pubertal, had preserved pulmonary

    function and suffered from pancreatic insuficiency. Each of these factors might have a

    major inluence on resting energy expenditure and growth40, but it seems reasonable

    to expect that these parameters were comparable between patients. In addition, we

    analyzed for possible inluence of several other parameters on the observed correlations

    in our multiple regression analysis (e.g. gender, age, mutation class). Yet, neither of

    these parameters seemed to affect our results.

    Since the CF nutritional recommendations still comprises high energy intakes for all

    pediatric CF patients, our data support the concept that sub-analysis of different groups

    of CF patients is necessary to categorize or even individualize the dietary advices. We

    believe it is important to investigate the relatively healthy CF patients with preserved

    pulmonary function, to conclude whether the actual beneicial effects on nutritional

    status or growth outweigh the possible adverse physiological consequences in our

    strive to achieve high dietary energy intakes. Our retrospective analysis in school-ageCF patients with preserved pulmonary function indicates no straight-forward relation

    between energy intake and nutritional status or growth. We do believe that our data

    supports the need for prospective nutritional studies in this subgroup of CF patients,

    but our results simultaneously underline the dificulty to design such nutritional trials

    with suficient statistical power.

    rEFErENCES

    1. Anthony H, Bines J, Phelan P, Paxton S (1998) Relation between dietary intake and nutritional

    status in cystic ibrosis. Arch Dis Child 78: 443-447

    2. Beker LT, Russek-Cohen E, Fink RJ (2001) Stature as a prognostic factor in cystic ibrosis

    survival. J Am Diet Assoc 101(4):438-442

    3. Cheng K, Ashby D, Smyth R (2000). Ursodeoxycholic acid for cystic ibrosis-related liver

    disease. Cochrane Database Syst Rev (2):CD000222

    4. Collins CE, OLoughlin EV, Henry RL (1997) Fat gram target to achieve high energy intake in

    cystic ibrosis. J Paediatr Child Health 31:142-147

    5. Constantini D, Padoan R, Curcio L, Giunta A (1988) The management of enzymatic therapyin cystic ibrosis patients by an individualized approach . J Pediatr Gastro Enterol Nutr 7

    (suppl):S36-S39

    6. Corey M, McLaughlin FJ, Williams M, Levison H (1988)A comparison of survival, growth, and

    pulmonary function in patients with cystic ibrosis in Boston and Toronto. J Clin Epidemiol

    41(6):583-591

    7. Duff AJ, Wolfe SP, Dickson C, Conway SP, Brownlee KG (2003) Feeding behavior problems

    in children with cystic ibrosis in the UK: prevalence and comparison with healthy controls. J

    Pediatr Gastroenterol Nutr 36(4):443-7

    8. Durie PR, Pencharz PB (1992) Cystic ibrosis: nutrition. Br Med Bull 48(4):823-846

  • 7/31/2019 thesis CF 4

    38/133

    38

    ChapTEr 2 INTaKE aNd GrOWTh

    9. Dutch Growth Foundation, growth and development in children. A programme for

    pediatricians and researchers, by pediatricians and researchers. The Netherlands 6 A.D.;

    2000-2006.

    10. Gangell CL, Hall GL, Stick SM, Sly PD (2010) Lung function testing in preschool-aged children

    with cystic ibrosis in the clinical setting. Pediatr Pulmonol 45:419-433

    11. Groeneweg M, Tan S, Boot AM, de Jongste JC, Bouquet J, Sinaasappel M (2002)Assessment

    of nutritional status in children with cystic ibrosis: conventional anthropometry and

    bioelectrical impedance analysis. A cross-sectional study in Dutch patients. J Cyst Fibros

    1(4):276-280

    12. Haeusler G, Frisch H, Waldhor T, Gtz M (1994) Perspectives of longitudinal growth in cystic

    ibrosis from birth to adult age. Eur J Pediatr 153(3):158-163

    13. Hanning RM, Blimkie CJR, Bar-Or O, Lands RC, Mos LA, Wilson WM (1993) Relationships

    among nutritional status and skeletal and respiratory muscle function in cystic ibrosis: does

    early dietary supplementation make a difference?Am J Clin Nutr 57:580-587

    14. Health Council of the Netherlands (2001). Dietary Reference Intakes: Energy, Proteins, Fats,and Digestible Carbohydrates Publication No. June 2002. Council of the Netherlands: The

    Hague, ISBN-10: 90-5549-384-8.

    15. Jelalian E, Stark LJ, Reynolds L, Seifer R (1998) Nutrition intervention for weight gain in

    cystic ibrosis: a meta analysis. J Pediatr 132(3 Pt 1):486-492.

    16. Kindstedt-Arfwidson K, Strandvik B (1988) Food intake in patients with cystic ibrosis on an

    ordinary diet. Scand J Gastroenterol Suppl 143:160-162

    17. Lenaerts C, Lapierre C, Patriquin H, Bureau N, Lepage G, Harel F et al (2003) Surveillance for

    cystic ibrosis-associated hepatobiliary disease: early ultrasound changes and predisposing

    factors. J Pediatr 143(3):343-350

    18. Lloyd-Still JD, Smith AE, Wessel HU (1989) Fat intake is low in cystic ibrosis despite

    unrestricted dietary practices. J Parenter Enteral Nutr 13:296-298

    19. Luder E, Kattan M, Thornton JC, Koehler KM, Bonforte RJ (1989) Eficacy of a nonrestricted

    fat diet in patients with cystic ibrosis. Am J Dis Child 143:458-464

    20. Mayer OH, Jawad AF, McDonough J, Allen J (2008) Lung function in 3-5 year old children with

    cystic ibrosis. Pediatr Pulmonol 43:1214-1223

    21. Patel L, Dixon M, David TJ (2003) Growth and growth charts in cystic ibrosis. J R Soc Med 96

    Suppl 43:35-41

    22. Powers SW, Byars KC, Mitchell MJ (2003) A randomized pilot study of behavorial treatment

    to increase calorie intake in toddlers with cystic ibrosis. Child Health Care 32(4):297-311

    23. Powers SW, Mitchell MJ, Patton SR, Byars KC, Jelalian E, Mulvihill MM, et al (2005) Mealtime

    behaviors in families of infants and toddlers with cystic ibrosis. J Cyst Fibros 4(3):175-82

    24. Powers SW, Patton SR, Byars KC, Mitchell MJ, Jelalian E, Mulvihill MM, et al (2002) Caloric

    intake and eating behavior in infants and toddlers with cystic ibrosis. Pediatrics 109(5):E75

    25. Powers SW, Patton SR (2003)A comparison of nutrient intake between infants and toddlers

    with and without cystic ibrosis. J Am Diet Assoc 103(12):1620-1625

    26. Ramsey BW, Farrell PM, Pencharz P (1992) Nutritional assessment and management incystic ibrosis: a consensus report. The Consensus Committee. Am J Clin Nutr 55(1):108-116

  • 7/31/2019 thesis CF 4

    39/133

    39

    ChapTEr 2 INTaKE aNd GrOWTh

    27. Shepherd RW, Holt TL, Cleghorn G, Ward LC, Isles A et al (1988) Short-term nutritional

    supplementation during management of pulmonary exacerbation in cystic ibrosis: a

    controlled study, including effects of protein turnover. Am J Clin Nutr 48:235-239

    28. Sinaasappel M, Stern M, Littlewood J, Wolfe S, Steinkamp G, Heijerman HG, et al (2002)

    Nutrition in patients with cystic ibrosis: a European Consensus. J Cyst Fibros 1(2):51-75

    29. Stallings VA, Stark LJ, Robinson MS, Feranchak AP, Quinton H (2008) Evidence-based practice

    recommendations for nutrition-related management of children and adults with cystic ibrosis

    and pancreatic insuficiency: Results of a systematic review. J Am Diet Assoc 108: 832-839

    30. Stark LJ, Bowen AM, Tyc VL, Evans S, Passero MA (1990)A behavorial approach to increasing

    calorie consumption in children with cystic ibrosis. J Pediatr Psychol 15(3):309-326

    31. Stark LJ, Knapp LG, Bowen AM, Powers SW, Jelalian E et al (1993) Increasing calorie

    consumption in children with cystic ibrosis: replication with 2-year follow up. J App Beh Anal

    26: 435-450

    32. Stark LJ, Opipari LC, Spieth LE, Jelalian E, Quittner AL et al (2003) Contribution of behaviour

    therapy to dietary treatment in cystic ibrosis: a randomized controlled study with 2-yearfollow-up. Behavior Therapy 34:237-258

    33. teinkamp G, Demmelmair H, Rhl-Bagheri I, von der Hardt, H, Koletzko, B (2000) Energy

    supplements rich in linoleic acid improve body weight and essential fatty acid status of cystic

    ibrosis patients. J Pediatr Gastro Enterol Nutr 31:418-423

    34. Stettler N, Kawchak DA, Boyle LL, Propert KJ, Scanlin TF, Stallings VA et al (2000)Prospective

    evaluation of growth, nutritional status, and body composition in children with cystic ibrosis.

    Am J Clin Nutr 72(2):407-413

    35. Tanner JM (1976) Clinical longitudinal standards for height, weight, height velocity, weight

    velocity, and stages of puberty. Arch Dis Child 51(3):170-179

    36. Vaisman N, Clarke C, Pencharz PB (1991) Nutritional rehabilitation increases resting energy

    expenditure without affecting protein turnover in patients with cystic ibrosis. J Pediatr

    Gastroenterol Nutr 13:383-390

    37. Vilozni D, Bentur L, Efrati O, Minuskin T, Barak A, Szeinberg A, et al (2007) Spirometry in

    early childhood in cystic ibrosis patients. Chest 131:356-361

    38. Walkowiak J, Przyslawski J (2003) Five year prospective analysis of dietary intake and clinical

    status in malnourished cystic ibrosis patients. J Hum Nutr Dietet 16:225-231

    39. Ward C, Massie J, Glazner J, Sheehan J, Canterford L, Armstrong D et al (2009) Problem

    behaviours and parenting in preschool children with cystic ibrosis. Arch Dis Child 94:341-347

    40. Wiskin AE, Davies JH, Wootton SA, Beattie RM (2010) Energy expenditure, nutrition and

    growth. Arch Dis Child 1-6

    41. Zielenski J, Tsui LC (1995) Cystic Fibrosis: Genotypic and phenotypic variations. Annu Rev

    Genet 29: 777-807.

  • 7/31/2019 thesis CF 4

    40/133

  • 7/31/2019 thesis CF 4

    41/133

    ChapTEr 3

    PERSISTENT FAT MALABSORPTION IN

    CYSTIC FIBROSIS;

    LESSONS FROM PATIENTS AND MICE

    M. Wouthuyzen-Bakker1

    F.A.J.A. Bodewes1

    H.J. Verkade1

    1Pediatric Gastroenterology, Department of Pediatrics, University Medical Center

    Groningen, University of Groningen, Groningen, The Netherlands.

    Jonl of Cystic Fibosis 2011; 10(3): 150-8

  • 7/31/2019 thesis CF 4

    42/133

    42

    ChapTEr 3 FaT MaLaBSOrpTION IN CYSTIC FIBrOSIS

    aBSTraCT

    Fat malabsorption in pancreatic insuficient cystic ibrosis (CF) patients is classically

    treated with pancreatic enzyme replacement therapy (PERT). Despite PERT, intestinal

    fat absorption remains insuficient in most CF patients. Several factors have been

    suggested to contribute to the persistent fat malabsorption in CF (CFPFM). We reviewed

    the current insights concerning the proposed causes of CFPFM and the corresponding

    intervention studies. Most data are obtained from studies in CF patients and CF mice.

    Based on the reviewed literature, we conclude that alterations in intestinal pH and

    intestinal mucosal abnormalities are most likely to contribute to CFPFM. The presently

    available data indicate that acid suppressive drugs and broad spectrum antibiotics could

    be helpful in individual CF patients for optimizing fat absorption and/or nutritional

    status.

  • 7/31/2019 thesis CF 4

    43/133

    43

    ChapTEr 3 FaT MaLaBSOrpTION IN CYSTIC FIBrOSIS

    INTrOduCTION

    Most patients with cystic ibrosis (CF) have a suboptimal nutritional status. In order to

    minimize morbidity and mortality, obtaining and maintaining a good nutritional status

    remains one of the important challenges in CF.1-3 Next to preventing and controlling

    pulmonary infections, a high dietary energy intake diet combined with pancreatic

    enzyme replacement therapy (PERT) are classic cornerstones in CF care.

    Despite optimizing and maximizing PERT, fat malabsorption in most CF patients persist

    (CFPFM). Rather than the physiological absorption above 95%, most CF patients show

    a decreased intestinal fat absorption of 85-90% of dietary fat intake.4,5 It is reasonable

    to assume that at least part of the failure to approach a physiological degree of fat

    absorption could still be attributed to ineficiency and inaccurate dosing of PERT. Also,

    a non-synchronous entrance of PERT with dietary energy intake into the duodenum

    may play a role in CFPFM.6 However, besides PERT related issues, other factors have

    been implied to be involved in CFPFM.7,8

    During the last decades, several of these potential contributing factors to CFPFM have

    been addressed and investigated in patient studies as well as animal studies. In this

    review, we will provide an overview of the different mechanisms that have been implied

    to contribute to CFPFM and describe the results of corresponding interventional studies

    on fat absorption and/or nutritional status. We will mainly describe studies performed

    in CF patients and CF mice. CF mice are especially suitable to exclusively investigate

    CFPFM, as most CF mouse models do not suffer from pancreatic insuficiency but do

    exhibit impaired absorption of fat.9 Based on the reviewed literature, we will concludeour review with clinical implications and suggestions for further research.

    In order to fully recognize and understand the CF-related factors that might be involved

    in CFPFM, we start to describe the physiological process of fat digestion and absorption.

    phYSIOLOGY OF FaT dIGESTION aNd aBSOrpTION

    Dietary fat intake mainly consists of long-, medium- and short-chain triglycerides. The

    mechanism of digestion and absorption of these lipids can be dissected into severalsequential steps. In contrast to long-chain triglycerides, medium- and short-chain

    triglycerides are known to circumvent several steps in lipid digestion and absorption.

    For example, unlike long-chain triglycerides, medium- and short chain triglycerides

    are less dependent upon solubilization, escape the re-esteriication into triglycerides

    and are directly absorbed into the portal system without being assembled into

    chylomicrons.10 Since the majority of dietary fat and energy intake consists of long-

    chain triglycerides (92-96%), we exclusively focused on their mechanism of intestinal

    digestion and absorption. The mechanism of digestion and absorption of long-chain

    triglycerides can be dissected into several sequential processes (igure 1A).

  • 7/31/2019 thesis CF 4

    44/133

    44

    ChapTEr 3 FaT MaLaBSOrpTION IN CYSTIC FIBrOSIS

    1) Emulsiication. Emulsiication of triglycerides is a process in which (water-insoluble)

    fat droplets are suspended in an aqueous environment. Emulsiication can be achieved by

    mechanical as well as chemical means. Mechanical emulsiication is attained by chewing

    and forcing dietary fat with high pressure through a small opening (e.g. the pylorus) and

    dispersing large fat droplets into smaller droplets. Chemical emulsiication is attained

    by the action of bile and prevents the emulsion from re-coalescing. Emulsiication

    results in a ine, relatively stable oil-in-water emulsion with an increased surface area.

    The water soluble digestive / lipolytic enzymes are active at the site of the water-oil

    interface.

    2) Lipolysis. During lipolysis, the breakdown (hydrolysis) of lipids into smaller particles

    is continued. Around 10-30% of the triglycerides are hydrolyzed in the stomach into

    diglycerides and free fatty acids, catalyzed by lingual and gastric lipase (a process also

    known as biochemical/lipolytic emulsiication10). Under physiological conditions,

    pancreatic lipase completes the process in the proximal part of the small intestine, byhydrolyzing the remaining triglycerides and diglycerides into monoglycerides and free

    fatty acid molecules. Bile salts are amphipathic molecules, i.e. they possess a hydrophilic

    and a hydrophobic site. Upon exposure to dietary lipid emulsion at the level of the

    proximal small intestine, the hydrophobic site orients itself towards the hydrophobic

    lipid droplets, whereas their hydrophilic site exposes itself to the aqueous (water) phase.

    Lipids droplets whose surface is thus covered bile salts are not accessible to pancreatic

    lipase. Pancreatic co-lipase allows the anchoring of the pancreas lipase to the oil-water

    interface and is essential for lipolytic activity. Under physiological conditions, pancreatic

    lipase is present in vast excess. Thus, in CF, fat maldigestion only occurs during severepancreatic malfunction.11 During exocrine pancreatic insuficiency when pancreatic

    lipase is severely reduced, lipolysis is more dependent upon the activity of lingual and

    gastric lipase. In CF conditions, lingual lipase remains fully active in the intestine, where

    it accounts for more than 90% of lipase activity, even when the intestinal pH drop below

    the optimal level for bile salt dependent lipolysis.80

    3) Solubilization. The process in which fat molecules are dissolved in water in the form of

    (mixed) micelles is called solubilization. Solubilization enhances the aqueous solubility

    of fatty acids by several orders of magnitude (100 to 1000 fold).10

    Solubilization isnecessary for monoglycerides and free fatty acids to eficiently overcome the diffusion

    barrier of the so called unstirred water layer of the enterocytes.10 The unstirred water

    layer separates the enterocytes from the luminal contents of the intestine. Solubilization

    is achieved by the formation of mixed micelles; mainly consisting of phospholipids and

    bile salts derived from biliary secretion. The diameter of the lipid droplets ranges from

    100 1000 nm, whereas that of mixed micelles ranges 3-5 nm. The hydrophobic part

    of the lipid molecules, s