The role of specific maternal nutritional factors on ...€¦ · on epigenetic programming of fetal...

155
The role of specific maternal nutritional factors on epigenetic programming of fetal immune development N. D. D. Manori Amarasekera MBBS, MPhil This thesis is presented for the degree of Doctor of Philosophy of University of Western Australia School of Paediatrics and Child Health 2015

Transcript of The role of specific maternal nutritional factors on ...€¦ · on epigenetic programming of fetal...

The role of specific maternal nutritional factors

on epigenetic programming of fetal immune

development

N. D. D. Manori Amarasekera MBBS, MPhil

This thesis is presented for the degree of Doctor of Philosophy of

University of Western Australia

School of Paediatrics and Child Health

2015

ii

DEDICATION

This thesis is dedicated to my mother, father and my sister who motivated me to go for

my highest potential and to Buddhika and Dehemi who continue to be my greatest

support and Nethmi for bearing up long hours of lab work with me in my womb for nine

months.

iii

STATEMENT OF CANDIDATE CONTRIBUTION

The author performed all the work presented in this thesis except where contributions

from others have been acknowledged below.

The work presented in this thesis was performed between February 2011 and December

2014 at the School of Paediatrics and Child Health, University of Western Australia

under the supervision of Professor Susan Prescott, Dr. Deborah Strickland, Dr. David

Martino and Professor Meri Tulic.

The Childhood Allergy and Immunology Research team (CAIR) assisted with

participant recruitment, data and blood collection, blood processing and data entry.

Professor Susan Prescott performed all clinical diagnoses.

Assistance with FACS was provided by Dr. Michelle Tourigny at Telethon Kids

Institute, Perth.

Dr. David Martino, Murdoch Children’s Research Institute, Melbourne (under the

supervision of Dr. Richard Saffery) was responsible for Illumina 450k methylation

array including designing and data analysis.

Hani Harb and Dr. Dorthe Kesper, Institute of Laboratory Medicine and

Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg, Germany

(under the supervision of Professor Harald Renz) performed and analysed the histone

acetylation experiments.

This work is original and has not been submitted previously for any other degree.

____________________ ____________________

Professor Susan Prescott N.D.D.Manori Amarasekera

Coordinating supervisor Candidate

iv

THESIS SUMMARY

The burden of non-communicable diseases (NCD) including cardiovascular, metabolic,

allergic and chronic lung diseases has reached pandemic proportions, as the major

global health challenge in the 21st century. Understanding the causes and mechanisms of

these diverse but pathogenically related inflammatory conditions remains a key

challenge in overcoming these trends. The rise in these NCD is strongly related to

modern lifestyle changes, and shared environmental risk factors, many of which disrupt

early metabolic immune programming and predisposed to subsequent disease.

The ‘Developmental Origin of Health and Disease’ (DOHaD) paradigm recognises the

crucial importance of the early environment in shaping both structural, developmental

and physiological response patterns in ways that may determine future health, biological

reserve and disease susceptibility. Among many environmental exposures, the quality of

early life nutrition is one of the most important factors influencing all aspects of

development during critical formative periods, particularly during gestation. While the

role of early metabolic adaptations has been long recognised as a predisposing factor,

the impact of the early environment on the developing immune system in the

subsequent predisposition to inflammation, has only been recognised more recently.

Identifying and optimising early factors that influence the patterns of immune

development is central in understanding disease pathogenesis and in implementing

better prevention strategies. In particular, variations in maternal nutrition can

significantly modify both immune and metabolic programming, to determine the long-

term health outcomes of the offspring. Only recently has it been recognised that many

of these effects are mediated through epigenetic mechanisms such as DNA methylation

and histone acetylation. The strong link between changes in the modern diet and

escalating rate of allergic and other NCD underscores the importance of identifying

specific in utero nutritional exposures affecting the fetal epigenome leading to an altered

immune profile.

The work of this thesis is amongst the first to explore the role of maternal nutrition on

neonatal epigenetic profile and subsequent risk for allergic outcomes using state-of-the-

art epigenomic profiling technologies.

v

The studies herein aimed to investigate epigenetic variation in neonatal immune cells in

association with variation in utero exposure to folate (chapters 3 and 4) and determined

whether maternal supplementation with fish oil containing omega-3 polyunsaturated

fatty acids (n-3 PUFA) modify the neonatal DNA methylome (chapter 5).

In study 1 (chapters 3 and 4), an ‘extremes of phenotype’ approach was employed to

retrospectively select a cohort of 23 cord blood samples from a prospective birth cohort

(n=420), categorised by high in utero folate (High Folate; HF, n=11) and low folate

(Low Folate; LF, n=12) exposure based on maternal serum folate levels measured in the

last trimester of pregnancy. DNA from both neonatal CD4+ T-cells and antigen

presenting cells (APC) isolated from cord blood were then bisulphite converted and

profiled for genome-wide methylation using Illumina Human Methylation 450 arrays.

In parallel, using a novel chromatin immunoprecipitation (ChIP) assay, histone 3 (H3)

and histone 4 (H4) acetylation levels at selected loci were measured in CD4+ T-cells as

an additional level of epigenetic regulation. A comparison of genome-wide DNA

methylation profiles between HF and LF groups revealed differential methylation at

seven regions across the genome. By far, the biggest effect observed was

hypomethylation of a 923bp region 3kb upstream of the ZFP57 transcript, regulator of

DNA methylation during development. Consistent differences in folate-sensitive

differentially methylated regions (fDMR) were reported in both CD4+ T-cells and APC,

which are derived from distinct hematopoietic cell lineages, suggesting these

differences have originated at the common hematopoietic progenitor stage. Additional

functional studies of mRNA expression and histone acetylation revealed that HF

exposures was associated with relatively higher mRNA expression and concomitant

higher H3 and H4 acetylation levels at the downstream ZFP57 gene. These findings

were replicated in an additional independent set of samples derived from the same

prospective birth cohort using Sequenom Mass Spectrometry and the findings from

these experiments were reported in FASEB medical journal.

Parallel studies of H3 and H4 acetylation levels at loci associated with allergy

development were undertaken to address a different hypothesis, was high folate

exposure is associated with variations in the activity of allergy-associated genes.. Using

targeted assays, these studies revealed significantly higher histone acetylation levels at

GATA3 locus, a master regulator of allergic immune responses, in neonates of the HF

group, suggesting increased propensity for activation of these pathways. In the same

vi

group a significant increase in H4 acetylation at the IL9 locus, another allergy-

promoting cytokine was observed. Together these findings suggest that exposure to high

gestational folate is associated with more transcriptionally permissive chromatin at

allergy-associated genes in CD4+ T-cells.

In study 2 (chapter 5) we studied the role of n-3 PUFA supplementation during

pregnancy, which previous studies in the host lab have shown to reduce the severity of

clinical symptoms in infants at high risk of allergic diseases. Experiments were carried

out to investigate whether these health promoting effects are mediated through DNA

methylation by analysing the CD4+ T-cells purified from cryo-preserved cord blood

samples selected from a randomised controlled trial of maternal fish oil

supplementation. In this double-blind trial, mothers were given either fish oil or placebo

from 20 weeks of pregnancy until delivery. Of the 80 mother-infant pairs who

completed the initial trial, cord blood samples of 70 neonates were available for

genome-wide DNA methylation profiling. Analysis of cord red cell fatty acid

parameters provided robust evidence that the maternal supplementation modified fetal

lipid profiles. There was very little evidence to support a role for DNA methylation in

mediating these effects. Comparison of DNA methylation profiles between treatment

and control groups did not reveal any statistically significant differences in CpG

methylation, at the single-CpG or regional level. Effect sizes for top-ranked candidates

were small and of unknown biological significance. Further studies would be required

to address the role of other epigenetic or post-transcriptional mechanisms that could

mediate these effects, however the experiments in chapter 5 argue against a substantial

role for DNA methylation.

Using cutting-edge epigenomic profiling technologies, this is the first study to

undertake extensive profiling of the neonatal epigenome in purified immune cells of

different lineages to unravel the combinatorial epigenetic effects of specific maternal

nutritional factors. This study provides evidence that epigenetic mechanisms play an

important role in mediating environmentally driven effects on neonatal genome. The

epigenomic regions identified in this thesis will provide the framework for future

studies in the evolving area of nutri-epigenomics to understand the origin of NCD and

help identify targets for treatment and prevention strategies.

vii

TABLE OF CONTENTS

DEDICATION............................................................................................................................ II

STATEMENT OF CANDIDATE CONTRIBUTION ........................................................... III

THESIS SUMMARY ................................................................................................................ IV

TABLE OF CONTENTS ....................................................................................................... VII

LIST OF FIGURES .................................................................................................................... X

LIST OF TABLES .................................................................................................................. XII

ABBREVIATIONS ................................................................................................................ XIII

DECLARATION OF PUBLISHED WORK ANDWORK PREPARED FOR

PUBLICATION ....................................................................................................................... XV

ACKNOWLEDGEMENTS ............................................................................................... XVIII

CHAPTER 1: LITERATURE REVIEW .................................................................................. 1

1.1 FETAL LIFE: THE CRITICAL PERIOD OF IMMUNE DEVELOPMENT ...................................... 1

1.1.1 Immunology of feto-maternal interface ...................................................................... 1

1.1.2 Ontogeny of immune development ............................................................................. 2

1.1.3 Transition of the immune profile from prenatal to postnatal life ................................ 3

1.1.4 Environmental exposures modulate immune responses: the effects of dietary factors

on immune development ...................................................................................................... 5

1.2 DEVELOPMENTAL PROGRAMING AND THE ‘DEVELOPMENTAL ORIGIN OF HEALTH AND

DISEASE (DOHAD)’ HYPOTHESIS ............................................................................................. 8

1.3 GENE-ENVIRONMENT INTERACTIONS IN DETERMINING DISEASE RISK ............................. 10

1.4 EPIGENETICS: WHAT IS IT? ................................................................................................ 11

1.4.1 Molecular mechanisms of epigenetics ...................................................................... 12

1.4.2 DNA methylation ...................................................................................................... 12

1.4.3 Histone modifications ............................................................................................... 14

1.4.4 Regulation by non-coding RNA (ncRNA) ................................................................ 15

1.4.5 Chromatin remodelling complexes ........................................................................... 16

1.5 FUNCTIONAL IMPLICATIONS OF EPIGENETIC DYNAMICS .................................................. 16

1.5.1 Epigenetic regulation during embryo development and haematopoietic development

............................................................................................................................................ 17

1.5.2 Epigenetic profile of the neonatal immune system and epigenetic changes during

postnatal development ........................................................................................................ 19

1.5.3 Evidence of disruption of epigenetic pathways in immune diseases......................... 20

viii

1.6 EPIGENETICS AS AN UNDERLYING MECHANISM OF DEVELOPMENTAL ORIGIN OF CHRONIC

DISEASES ................................................................................................................................ 22

1.7 MATERNAL ENVIRONMENTAL EXPOSURES THAT MODULATE THE NEONATAL EPIGENOME

................................................................................................................................................ 23

1.7.1 Nutritional exposures in early life and the fetal epigenome ...................................... 23

1.7.1.1 Folate and other nutrients involved in one-carbon metabolism ......................... 24

1.7.1.2 Polyunsaturated fatty acids (PUFA) .................................................................. 29

1.7.2 Other maternal exposures that affect the fetal epigenome ........................................ 30

1.8 CHALLENGES OF EPIGENOME-WIDE ASSOCIATION STUDIES ............................................. 33

1.9 CONCLUDING REMARKS .................................................................................................... 34

CHAPTER 2: STUDY RATIONALE, STUDY DESIGN AND METHODS ...................... 36

2.1 STUDY RATIONALE ............................................................................................................ 36

2.2 STUDY COHORTS ............................................................................................................... 37

2.3 DATA AND SAMPLE COLLECTION ...................................................................................... 38

2.3.1 Questionnaire data ..................................................................................................... 38

2.3.2 Maternal and infant skin prick test (SPT) ................................................................. 38

2.3.3 Clinical assessment for allergic diseases ................................................................... 39

2.3.4 Collection of cord and maternal peripheral blood ..................................................... 39

2.4 LABORATORY METHODS ................................................................................................... 39

2.4.1 Harvesting and cryopreservation of mononuclear cells ............................................ 39

2.4.2 Thawing of cryopreserved mononuclear cells ........................................................... 40

2.4.3 Cord blood immune responses .................................................................................. 41

2.4.4 Purification of CD4+ T-cells ...................................................................................... 41

2.4.5 Purification of antigen presenting cells ..................................................................... 42

2.4.6 Purification of genomic DNA and total RNA ........................................................... 43

2.4.7 Quantitative polymerase chain reaction (qPCR) ....................................................... 43

2.4.8 DNA methylation analysis ........................................................................................ 45

2.4.9 Validation of methylation array targets ..................................................................... 45

2.4.10 Histone 3 and Histone 4 acetylation analysis .......................................................... 46

2.5 STATISTICAL ANALYSIS .................................................................................................... 46

2.6 ETHICS APPROVAL ............................................................................................................ 46

CHAPTER 3: PAPER 1 ........................................................................................................... 47

3.1 ABSTRACT ......................................................................................................................... 48

3.2 INTRODUCTION ................................................................................................................. 49

3.3 MATERIALS AND METHODS .............................................................................................. 50

3.4 RESULTS ............................................................................................................................ 54

3.5 DISCUSSION ....................................................................................................................... 62

ix

3.6 REFERENCES ..................................................................................................................... 64

CHAPTER 4: PAPER 2 ........................................................................................................... 67

4.1 ABSTRACT ......................................................................................................................... 68

4.2 INTRODUCTION ................................................................................................................. 69

4.3 METHODS .......................................................................................................................... 70

4.4 RESULTS ............................................................................................................................ 73

4.5 DISCUSSION ....................................................................................................................... 77

4.6 REFERENCES ..................................................................................................................... 79

CHAPTER 5: PAPER 3 ........................................................................................................... 81

5.1 ABSTRACT ......................................................................................................................... 82

5.2 INTRODUCTION ................................................................................................................. 83

5.3 MATERIALS AND METHODS .............................................................................................. 84

5.4 RESULTS ............................................................................................................................ 86

5.5 DISCUSSION ....................................................................................................................... 90

5.6 REFERENCES ..................................................................................................................... 93

CHAPTER 6: GENERAL DISCUSSION ............................................................................... 96

6.1 DISCUSSION OF KEY FINDINGS IN THE CONTEXT OF EXISTING LITERATURE ..................... 96

6.2 LIMITATIONS OF THE STUDY ........................................................................................... 100

6.3 DIRECTIONS FOR FUTURE STUDIES IN THIS AREA ........................................................... 102

6.4 CONCLUDING REMARKS .................................................................................................. 103

BIBLIOGRAPHY ................................................................................................................... 105

APPENDIX 1 ........................................................................................................................... 133

APPENDIX 2 ........................................................................................................................... 134

APPENDIX 3 ........................................................................................................................... 137

x

LIST OF FIGURES

Figure 1.1 Schematic representation of the organisation and packaging of nucleosome Figure 1.2 Schematic illustration of one carbon metabolism Figure 2.1 Outline of purification steps used to isolate CD4+ T-cells and antigen presenting cells (APC) from cord blood mononuclear cells (CBMC) for the studies in this thesis Figure 2.2 Outline of screening strategy for isolating antigen presenting cells (APC) using FACS analysis Figure 3.1 Relationship between maternal serum folate and cord serum folate levels Figure 3.2 Differential methylation analysis between CD4+ and APC cells Figure 3.3 Analysis of global patterns of DNA methylation between folate groups Figure 3.4 Folate sensitive differentially methylated region (fDMR) in chromosome 6 Figure 3.5 This figure shows validation data of chromosome 6 fDMR and functional assessment of ZFP57 in CD4+ cells Figure 3.6 Sequenom Epityper analysis of DNA methylation at the chromosome 6 fDMR in an independent set of samples (n=19) Figure 4.1 Outline of the ChIP assay and shearing efficiency Figure 4.2 Lower limit of quantification and freeze-thawing effect of chromatin Figure 4.3 Levels of H3 (A) and H4 (B) histone acetylation at key genes associated with allergic inflammation in neonatal CD4+ T-cells Figure 5.1 Cord red blood cell fatty acid measurements in fish oil and control groups Figure 5.2 Regression analysis of CpG sites that co-associate with cord red cell total n-3 fatty acid levels

12 25 41 43 54 56 57 58 59 60 73 74 75 87 89

xi

Figure 5.3 Comparative analysis of DNA methylation profiles between treatment and intervention groups

89

xii

LIST OF TABLES

Table 1.1 Gene loci that show epigenetic modulation in association

with immune diseases

Table 3.1 Population characteristics of the mother and infants in

the discovery cohort

Table 3.2 Differentially methylated regions (DMR) according to

the folate status

Table 4.1 The effect of temperature and long-term storage on per

cent enrichment of H3 and H4 histone acetylation ± SEM

Table 5.1 Population characteristics of the mothers and infants in

the cohort (n=70)

21

55

58

74

86

xiii

ABBREVIATIONS

2ME 2-mercaptoethanol

5hmC 5-hydroxymethylcytosine

5mC 5-methylcytosine

AHRR aryl hydrocarbon receptor repressor

APC antigen presenting cells

bp base pairs

CBMC cord blood mononuclear cells

cDNA complementary DNA

ChIP chromatin immunoprecipitation

CpG cytosine followed by a guanine, linked together by a phosphate

DC dendritic cells

DEP diesel exhaust particles

DHA docosahexanoic acid

DMP differentially methylated positions

DMR differentially methylated region

DMSO dimethyl sulphoxide

DNMT DNA methyltransferases

DOHaD developmental origin of health and disease

EPA eicosapentaenoic acid

EWAS epigenome-wide association studies

FCS fetal calf serum

fDMR folate sensitive-differentially methylated regions

FOXP3 forkhead box 3

GATA3 GATA binding protein 3

gDNA genomic DNA

GWAS genome-wide association studies

H3 histone 3

H3K4me/me2/me3 mono/di/tri-methylation of lysine 4 of Histone 3

H3Kn lysine (n) of Histone 3

H4 histone 4

HAT histone acetyltransferases

HF high folate

HIFCS heat-inactivated fetal calf serum

xiv

HKG house keeping gene

IFNG interferon gamma

IGF2 insulin-like growth factor-2

IL- interleukin

IP mmunoprecipitation

LCPUFA long chain polyunsaturated fatty acids

LF low folate

MBD methyl binding domain

MC mononuclear cells

MDS multidimensional scaling

miRISC miRNA-associated RNA-induced silencing complex

miRNA microRNA

n-3 PUFA omega 3 polyunsaturated fatty acids

NCD non-communicable diseases

ncRNA non coding RNA

NIMA non-inherited maternal alloantigens

OVA ovalbumin

PCR polymerase chain reaction

PGC primordial germ cells

PM particulate matter

qPCR quantitative polymerase chain reaction

RCT randomised controlled trial

RPMI Roswell Park Memorial Institute

RT room temperature

SAM S-adenosylmethionine

SCFA short chain fatty acids

SDS sodium dodecyl sulfate

SNPs single nucleotide polymorphisms

SPT skin prick test

TET ten-eleven translocation proteins

Th1/2 T helper1/2

Tregs regulatory T-cells

TSLP thymic stromal lymphopoietin

UTR untranslated region

xv

DECLARATION OF PUBLISHED WORK AND

WORK PREPARED FOR PUBLICATION

This thesis contains published work and work prepared for publication, all of which has

been co-authored. The bibliographical details of the work, where it appears in the thesis

and author contributions are outlined below.

1. Genome-wide DNA methylation profiling identifies a folate-sensitive region of

differential methylation upstream of ZFP57 imprinting regulator in humans

Manori Amarasekera, David Martino, Sarah Ashley, Hani Harb, Dörthe Kesper,

Deborah Strickland, Richard Saffery, and Susan L. Prescott

Published in: FASEB J. 2014 Jun 2. pii: fj.13-249029. [Epub ahead of print] and

constitutes Chapter 3 of the thesis.

M. Amarasekera was involved in study conception and design, isolation of CD4+ T-

cells, sorting APC, DNA extraction, qPCR assay, target gene validation for methylation,

data analysis and manuscript preparation. All authors assisted in editing the manuscript

content and approved the final manuscript.

2. Folate status as a modifier of H3/H4 histone acetylation marks at allergy-

associated genes in human neonatal CD4+ T-cells

Hani Harb* Manori Amarasekera*, Sarah Ashley, Meri K. Tulic, Petra Ina Pfefferle,

Daniel P. Potaczek , Harald Renz , David Martino, Dörthe A. Kesper & Susan L.

Prescott

*HH and MA contributed equally to this work and share first authorship.

In preparation to be submitted to Epigenetics and constitutes Chapter 4 of the thesis.

M. Amarasekera was involved in study conception and design, isolation of CD4+ T-

cells, qPCR assay, cell culture experiments, cytokine analysis, data analysis and

manuscript preparation. All authors assisted in editing the manuscript content and

approved the final manuscript.

xvi

3. Epigenome-wide analysis of neonatal CD4+ T-cell DNA methylation sites

potentially affected by maternal fish oil supplementation

Manori Amarasekera, Paul Noakes, Deborah Strickland, Richard Saffery, David J

Martino, Susan L Prescott

Published in: Epigenetics. 2014 Dec 7:0. [Epub ahead of print] and constitutes Chapter

5 of the thesis.

M. Amarasekera assisted in study conception and design, isolation of CD4+ T cells,

flow cytometry analysis, DNA extraction, data analysis and manuscript preparation. All

authors assisted in editing the manuscript content and approved the final manuscript.

4. Nutrition in early life, immune-programming and allergies: the role of

epigenetics

Manori Amarasekera, Susan L. Prescott and Debra J. Palmer

Published in: Asian Pac J Allergy Immunol. 2013 Sep;31(3):175-82.

M. Amarasekera drafted and assisted in editing the manuscript.

5. Epigenetic aberrations in human allergic diseases

Manori Amarasekera, David Martino, Meri K Tulic, Richard Saffery and Susan

Prescott

Published in: Tryve O. Tollesfsbol, editor. Epigenetics in human disease. Philadelphia:

Elsevier Inc. 2012. p371-387.

M. Amarasekera drafted and assisted in editing the manuscript.

6. Epigenetics in immune development and in allergic and autoimmune diseases

David Martino, Dorthe A. Kesper, Manori Amarasekera, Hani Harb, Harald Renz and

Susan Prescott.

xvii

Published in: J Reprod Immunol. 2014 Oct;104-105:43-8.

M. Amarasekera assisted in drafting and editing the manuscript.

____________________ ____________________

Professor Susan Prescott N.D.D.Manori Amarasekera

Coordinating supervisor Candidate

xviii

ACKNOWLEDGEMENTS

I would like to thank the study participants and the obstetricians and midwives who

dedicated their time and effort to make this study possible.

My sincere gratitude and eternal thanks go to my primary supervisor Professor Susan

Prescott for offering me the opportunity to work on this project. You motivated me to

believe in myself and in my potentials. Thank you for the guidance and generous

opportunities along the way. I am very grateful also to Dr. Deborah Strickland and Prof.

Meri Tulic, my co- supervisors, for the time, advice and support. Many thanks to Dr.

David Martino for being so patient with me. I could always ring you to get advice in

difficult situations. Without your support and advice I would not have been able to

perform complex molecular work involved in this thesis.

Many thanks go to Dr. Anthony Kicic, Dr. Erika Sutanto and Thomas Iosifidis for their

excellent advice and assistance with qPCR. Thanks to Nina D’Vaz for her advice and

assistance with Luminex protein assays. I would like to thank Dr. Michelle Tourigny for

her assistance in FACS. Thanks to the Cancer and Disease Epigenetic Group, Murdoch

Children’s Research Institute, Melbourne for their assistance in Sequenom work and Dr.

Richard Saffery for his advice on methylation data analysis and interpretation. I would

also like to thank the Childhood Allergy and Immunology Research Group – Debra

Palmer, Paul Noakes, Suzanne Meldrum, Rachel West, Sarah Miller, Catherine Haigh,

Emma Prescott and Emma Snelgar. Thanks go to my fellow PhD students – Alexandra

Heaton, Valene See, Jessica Metcalfe and Anderson Jones for your support.

I would also like to acknowledge the financial support I received from the Australian

Government (Endeavour Postgraduate scholarship).

Finally I would like to thank my husband and my two gorgeous daughters for being

there for me as a constant source of strength.

1

Chapter 1

Literature Review

This literature review examines evidence for in utero epigenetic variations as mediators

of environment-induced changes that may modify later health outcomes, particularly

immune diseases. As a prelude to this, the initial sections of this review briefly

summarise the ontogeny of the immune system, how epigenetics are involved in

immune development, and how disruptions in epigenetic regulation are associated with

disease states.

1.1 Fetal life: the critical period of immune development

There has been a significant increase in immune-mediated diseases over the past 4-6

decades (1). This has been clearly associated with the marked environmental changes

associated with transition to more modern life-styles (2). The dramatic rise in infant

immune diseases, most notably allergy, indicates the specific vulnerability of the

immune system to early environmental changes. The associated parallel rise in

metabolic diseases that are characterised by low-grade inflammation, including obesity,

childhood type 2-diabetes and non-alcoholic fatty liver disease (collectively known as

non-communicable diseases, NCD) further highlights the importance of the finely

balanced development of the immune system (2, 3). Allergic diseases frequently

manifest within the first months of life (4) and this strongly suggests that the developing

immune system is exquisitely sensitive to environmental changes and that events

occurring during pregnancy play a central role in determining immune development.

1.1.1 Immunology of feto-maternal interface

The mammalian fetus can be considered as a highly successful allograft. The placenta is

the immunologically active interface coordinating fetal and maternal immune systems to

coexist during pregnancy (5). The placental cytokine milieu (higher levels of IL-4, and

IL-5) and hormonal environment (progesterone and prostaglandin E2) directs adaptation

of cellular and humoral immune response at the feto-maternal interface to favour

maternal tolerance to allogenic fetal cells (6-8). Furthermore, there is also expansion of

maternal regulatory T-cells (Tregs) during pregnancy, and these are also recruited to the

feto-maternal interface where they orchestrate immune tolerance to the fetus (9). There

2

is accumulating evidence that epigenetic mechanisms (section 1.5.2) provide the

biological mechanisms underpinning the establishment and maintenance of the distinct

immunological environment that is essential for optimal fetal development. As the

primary interface between maternal and fetal circulations, placenta is potentially

exposed to a wide range of environmental factors through maternal exposures. There is

evidence that maternal factors influence the placental function and affect the fetal

immune and metabolic development, indicating the importance of intrauterine

environment in shaping the immune development. (10-12).

1.1.2 Ontogeny of immune development

The fetal immune system was long considered as an immature version of the adult

immune system (13), however, Mold and McCune recently challenged this notion (14).

While many aspects of fetal immune responses are immature, there is now evidence of

more active immune regulation and subtle complexities that are adaptive for the unique

environment of the fetus. Several lines of evidence suggest that development of the

mammalian immune system occurs in distinct layers, with unique functions at different

stages, rather than a simple progression from an ‘immature state’ to a ‘mature state’ in a

linear fashion (14). This “layered immune hypothesis” has been supported by

observations from animal models that revealed the existence of unique waves of

lymphocyte production during fetal and neonatal development. In a chick chimeric

model, it was initially observed that populations of thymocytes with different

characteristics appear in the thymus at different stages of fetal development, completely

replacing earlier populations (15). This suggests that functionally different cell types are

acquired during different stages of development. Subsequently, these observations were

extended to murine models providing further evidence that the immune system develops

in stratified layers (16-18).

In humans, Mold and colleagues recently revealed similar developmental processes in

ontogeny of human haematopoiesis. Notably, they found that fetal T-cells show a

stronger developmental bias towards tolerance compared to adult T-cells. This is likely

to reflect the need to develop an initial broad repertoire of regulatory T-cells that are

tolerant to self-antigens and other newly encountered harmless antigens (19). Thus, a

more tolerogenic fetal immune system provides survival benefit in preventing a

deleterious response to non-inherited maternal alloantigens (NIMA) that can be passed

to the fetus (19, 20). Nevertheless, there is much to be explored as to how and when

3

these developmental variants occur and what triggers the switch from one phase to the

other. It is intriguing to explore whether the tolerogenic properties of fetal immunity

extend beyond self-antigens and NIMA to foreign antigens, since it may have both

beneficial and detrimental consequences. Detailed examination of human

haematopoiesis is beyond the scope of this thesis, however, studies further exploring

fetal immune development will have the capacity to fill the gaps in our understanding of

pathogenesis of NCD including allergic diseases.

1.1.3 Transition of the immune profile from prenatal to postnatal life

The active ‘tolerogenic’ state of the fetal immune system is maintained through a

complex and coordinated network of immune cells (14, 19, 21). While maintaining

immunological ‘quiescent’ state, this active surveillance network also appears to shape

the fetal antigen-specific effector responses to promote fetal survival (22, 23) by

providing an appropriate level of immune defence while limiting excessive effector

responses to self-antigens and NIMA. Fetal dendritic cells (DC) and Tregs provide

signals directing T-helper (Th) cell differentiation along the Type-2 helper (Th2)

pathway, away from potentially destructive Type-1 helper (Th1) responses (21-23).

This Th2 / regulatory-biased immune environment is observed both in the peripheral

immune system (24, 25) as well as in the thymic microenvironment (26) highlighting

the tight and coordinated regulation of activation-induced immune responses at multiple

levels to confer survival benefits.

A large body of data has demonstrated differences in immune function at birth in

neonates who subsequently develop allergic disease, indicating in utero events are

playing a critical role in shaping the fetal immune development (4). Allergy-prone

neonates demonstrate altered functions in adaptive (24), innate (27-29) and Tregs (26,

30) compartments suggesting disease result from disruption to an extensive immune

network rather than from a defect in an isolated cell subset. Reduced capacity for Th1

function and Th2-skewed allergen-specific responses are prominent in high risk

neonates (24). As further discussed below a number of maternal factors such as

maternal dietary nutrients, microbial exposure and exposure to pollutants have shown to

modify immune profiles at birth (31). While the origin and the functional relevance of

specific immune responses to exogenous antigens including allergens during fetal life

are not clear, it highlights, together with recognised associations of maternal exposures

4

with risk of developing allergic disease (2), that in utero exposures can have a greater

impact on immune development.

With the transition to the postnatal environment the developing immune system faces

another series of challenges, as the infant encounters a microbe-rich extra-uterine

environment. The immune system must establish balanced mutualism with colonising

commensals, while maintaining defence against potential pathogens. As such, the

immediate postnatal period is another crucial time for the induction of an expanded

repertoire of specific immunological tolerance to a wider range of nearly encountered

harmless antigens, and a broad repertoire of memory responses to potential pathogens

(32). To tailor these functional requirements of postnatal life, there is a gradual “shift”

of the default responses over the first years of life. One of the most obvious transitions

is from a relatively Th2 dominant default response in the perinatal period, to a more

Th1 dominated responses over the preschool years (24, 27) to eventually achieve the

‘mature’ patterns of response observed in adult life. As part of this process, adaptive

changes can be observed to occur in innate (22, 27, 33, 34) and Treg function (26, 35)

and the patterns of effector T-cell responses (24, 36), to achieve functionally distinct

immune cells that converge to meet the developmental stage-appropriate requirements.

Early interaction with the microbial environment is arguably the most critical factor

driving normal maturation of the immune system in favour of increasing Th1 propensity

as well as mature regulatory function (1). The likely importance of microbial exposure

is demonstrated in animal models which show grossly abnormal development of the

intestinal immune systems of mice raised in germ-free environments (37). There is also

evidence in humans that altered patterns of gut microbiota in the early postnatal period

are associated with dysregulated immune development in childhood (38, 39) and that

reduced biodiversity of gut microflora in the first months of life is associated with

subsequent allergic outcomes (32, 38, 39).

In summary, evidence suggests that immune development evolves in distinct functional

waves that serve different purposes at different developmental stages. During this

process, fetal and early postnatal period represent critical periods that are susceptible to

modulation. Nutritional patterns and a plethora of other early environmental factors

influence this immune development and maturation both by modulating the microbiota

and through direct immunomodulating effects. As discussed further below, some of

5

these mediate their effects by modulating the epigenome (described in detail in section

1.7).

1.1.4 Environmental exposures modulate immune responses: the effects of dietary

factors on immune development

Complex environmental and lifestyle changes are implicated in the dramatic increase in

NCD (2). Of these, modern dietary changes are among the most likely environmental

culprits implicated in the rising risk of so many diverse NCD (3, 40). Although there are

wide regional variations, many modern diets typically contain more processed and

synthetic foods rich in fats and refined carbohydrates with lower amounts of fibre, fresh

fish, fruits and vegetables compared to more traditional diets. These changes have been

associated with changes in the gut microbiome, metabolic responses and immune

function - all of which may contribute to chronic low-grade inflammation and altered

homeostatic mechanisms which are common risk factors for virtually all NCD.

Epidemiological studies have revealed how favourable dietary patterns, such as the

Mediterranean diet in pregnancy and in early childhood can have a protective effect on

persistent wheeze and atopy in children (41, 42). This dietary pattern is also associated

with reduced risk of diabetes (43), cardiovascular disease, some cancers and other NCD

(44). Notably, these beneficial effects reflect composite dietary patterns, and are

difficult to attribute to a single dietary element. On the other hand, there are also a

number of studies that have taken a ‘component’ approach to demonstrate the specific

immunomodulatory properties of individual dietary components such as vitamins,

minerals, short chain fatty acids (SCFA), and long-chain polyunsaturated fatty acids

(LCPUFA) (40). A key aim of this thesis was to explore the role of key dietary

components identified as risk factors in these studies, and to examine their potential

effects on the developing epigenome.

The local tissue microenvironment is an important determinant of immune

development. Changes in the local tissue milieu can modify the pattern of effector

response (45). Environmental factors which modify the local microenvironment,

therefore, have significant potential to alter immune development and the propensity for

inflammation. Of these, a range of dietary factors (such as LCPUFA and antioxidants)

have recognised effects on tissue milieu as a result of their influence as metabolic

components, substrates or structural components of cells and tissues, with downstream

effects on gene expression through a number of different pathways (46, 47). LCPUFA

6

are good examples of dietary factors that have multiple metabolic, immune and

structural functions that influence the propensity for inflammation. As structural

components of cell membranes, they influence membrane fluidity and cell signalling

(48). As substrates for prostanoid production, they influence the level of inflammatory

prostaglandins (49) and as substrates for resolvins they influence the local control of

inflammation (50). Animal studies demonstrate that dietary modulation with LCPUFA

(a combination of omega-3; n-3, and omega-6; n-6, PUFA) can change the local

production of immunomodulatory factors by the skin keratinocytes including IL-10 and

thymic stromal lymphopoietin (TSLP) thereby influencing skin inflammation (51). In

humans, it has been demonstrated that high dose n-3 PUFA supplementation in

pregnancy can modulate fetal oxidative stress (52), leukotriene metabolism (53) with

associated effects on immune function in cord blood (54). Clinical trials in pregnancy

and during the early postnatal period using fish oil supplementation have shown that

these immunomodulatory properties of n-3 PUFA have been associated with a reduction

in some allergic outcomes suggesting that the biological effects have clinical relevance

(54-57). This adds credence to the epidemiological observations that reduced intake of

n-3 PUFA and increased intake of nutrients rich in n-6 PUFA such as margarine has

shown to be associated with the rise in allergic disease (58-60). Section 1.7.1.2 of this

thesis explores the role of n-3 PUFA supplementation during pregnancy and its effects

on the epigenome during fetal immune development.

Antioxidants (such as selenium, zinc, vitamin C and vitamin E) are other examples of

dietary nutrients that have immunomodulatory effects potentially through changes in the

local tissue milieu. Studies using isolated human cells have shown that by favourably

altering the ‘redox’ status of cells, antioxidants can enhance IL-12 production by

antigen presenting cells (APC) to promote Th1 differentiation (61), although it is not

clear if this can be extrapolated to the in vivo setting. Observational studies suggest that

higher dietary intakes of antioxidant rich foods (such as fresh fruits and vegetables) or

higher antioxidant levels measured in pregnancy (62, 63) and early childhood (64, 65)

may reduce the risk of wheezing, asthma and/or eczema. As yet, there are no

intervention studies in early life to directly examine potential preventive effects. In part

this has been because there are two contrary hypotheses around the role of dietary

antioxidant intake and immune outcomes (66). While antioxidants have been suggested

to protect against allergic disease, there has also been conjecture that oxidative stress,

which increases the production of reactive oxygen species by macrophages could favour

7

Th1 immune differentiation. This alternative hypothesis proposes a theoretical concern

that antioxidant supplementation could increase the probability of Th2 differentiation

(by inhibiting oxidative stress) and favour the development of asthma and allergic

disease (67).

Changing dietary patterns are also implicated in altered microbiota. While this was

initially attributed to more ‘hygienic environments’, antibiotic use (68, 69) and reduced

exposure to infectious agents (70), it is now recognised that dietary profiles are also a

major determinant of the gut microbiome and biodiversity (71). There is now good

evidence that high-fat, low-fibre diets can alter the gut microbiome promoting low-

grade chronic inflammation (72, 73). Experimental mouse models have been used to

demonstrate the importance of neonatal colonisation with a diversified intestinal

microbiota for successful induction of oral tolerance to ovalbumin (OVA) (74, 75).

They also demonstrate the beneficial effects of dietary fibre and SCFA which are by-

products of fermentation of dietary fibre, in modulating the microbiome and anti-

inflammatory effects (76-78). In humans, use of prebiotic fibre to promote more

favourable colonisation in infancy has been associated with demonstrable reduction in

allergic disease (77, 79), underscoring the role of dietary nutrients in modulating

immune development through effects on the intestinal microbiota.

There are also interactions between metabolic and immune pathways and the gut

microbiota. The changes in gut immune regulation implicated in the rise in allergy and

autoimmunity (38, 39) have also been linked to the rise in obesity and metabolic disease

(80, 81). The changes in the gut microbiome induced by a high-fat, low-fibre diets have

been associated with changes in gut permeability, higher systemic antigenic load (low

grade endotoxaemia) and higher serum levels of inflammatory cytokines (72, 73).

Animal studies demonstrate how gut microbiota can regulate expression of genes that

affect fatty acid oxidation and fat deposition in host adipocytes (82). In humans, obese

individuals have documented differences in gut microbiota compared to lean

individuals, suggesting a role of gut microbiota in maintaining host metabolic

homeostasis (83). These observations collectively highlight the complex interplay

between metabolic and immune programming in the gut and provide new perspectives

on how diet-induced alterations to gut microbiome can have multisystem effects.

8

A range of other specific nutrients have also been implicated in the rise of allergic

conditions and many other inflammatory diseases (40). Some of these, such as vitamin

D, have recognised effects on immune function, as well as epidemiological associations

with allergic disease and other NCD (84). Others, such as folate, have been of particular

interest because of epigenetic effects on immune programming in animal models

(below). Collectively these studies underscore the importance of early events during

immune programming and how dietary factors can influence these processes. This is a

key theme of the current thesis, and is explored in detail in chapters 3, 4 and 5.

1.2 Developmental programing and the ‘Developmental Origin of Health and

Disease (DOHaD)’ hypothesis

Extensive data from both human and animal studies indicate that mammalian

development involves a number of periods of critical “programing” that shapes

physiologic and metabolic functions in ways that determines adaptive responses and

resilience to subsequent challenges, and susceptibility to disease in later life (85). While

these adaptations may be to promote optimal development, or ensure survival under

adverse uterine conditions, some of these effects may have deleterious effects on long-

term health increasing vulnerability to disease in later life (86).

Barker and colleagues were the first to highlight this phenomenon by demonstrating

nutritional patterns in early life are linked to the risk of cardio-vascular and metabolic

diseases many decades later (86, 87). Barker referred this to the ‘fetal origins of chronic

diseases’ hypothesis (87) but it is now recognised that these influences extend before

conception and also developmental plasticity continues into the postnatal period, as

reflected in the now broader nomenclature of the “Developmental Origin of Health and

Disease’ (DOHaD) (88). The notions of DOHaD established through this new field of

research, have led to more cross-disciplinary approaches to investigate mechanisms

underpinning developmental programming of many inter-related organ systems, which

are influenced by many common risk factors in early life (87, 89).

The original observations that formed the basis of the DOHaD hypothesis were centred

around the effects of maternal under-nutrition at critical stages in early life, leading to

persisting physiological adaptation as well as growth patterns. Barker’s observation led

to the realisation that some consequences may only become apparent later in adult life.

Specifically, a series of observational studies conducted in United Kingdom found that

9

low birth weight, as a crude marker of fetal under-nutrition was associated with greater

risk of heart disease, stroke, hypertension and diabetes in later life (87). These findings

have been since supported by animal experiments using dietary manipulation to

replicate the effects of a wide variety of early risk factors associated with metabolic and

cardiovascular disease (90-93). While DOHaD hypothesis has provided logical

explanation for the growing epidemiological literature on associations between early-

life exposures and later health status, “predictive adaptive response’ hypothesis that was

proposed by Bateson and Gluckman (94) has attracted attention of the epidemiologists

working in the DOHaD field. It posits that the developing organism have the capacity

to receive the information about the quality of the external environment, and in

response, formulate predictions as to future ecological conditions. When the predicted

and actual environments differ, the mismatch between the individual's phenotype and

the conditions in which it finds itself can have adverse consequences for later health.

Observations in animals have supported this model (95), however, it is not clear how

relevant is this in humans.

More recently, chronic-low grade inflammation has been recognised as a key element in

both the initiation and perpetuation of cardiovascular, metabolic and immune diseases.

This has driven interest in early environmental factors that may determine the dynamics

of inflammation in adulthood. In this context, McDade and colleagues found that low

birth weight, in addition to effects on metabolic dysregulation, is also an independent

risk factor for elevated baseline C-reactive protein (as a measure of low-grade systemic

inflammation) in adulthood (96). This suggested early effects on immune development

as well as metabolic pathways, providing additional mechanisms for the observed

associations between adverse early life conditions and increased incidence of

cardiovascular and other NCD in later life. Chronic low-grade inflammation as a

characteristic feature of virtually all NCD, also highlights the central role of the immune

system in the risk and pathogenesis of NCD, and further underscores the need to

investigate early life origin of immune disease and immune dysregulation.

In this context, the recent epidemic of allergic diseases takes on new significance.

Allergic conditions are typically the earliest onset NCD often presenting within the first

months of life (4), as a clear early indicator of the rising propensity for inflammation

and immune dysregulation and the very early vulnerability to modern environmental

changes. Many studies now show significant differences in the neonatal immune

10

function of children who subsequently develop allergy, adding to evidence that allergic

disease also has origins in utero. This is reflected in well-documented differences in

adaptive (24, 97-99) and innate immune compartments (27) as well as regulatory

networks (10, 26) in allergic children at birth. Many of these studies suggest relative

immaturities of T-cell function compared to non-allergic children. Consistent with this,

in a recent study, T-cell signalling pathways showed to be differently expressed at birth

between allergic and non-allergic children with marked down regulation of NF-кB

genes in allergic individuals (100). Furthermore, many risk factors for other NCD

(microbial exposures, diet and pollutants), also have been associated with differences in

immune function at birth, also suggesting in utero effects (40, 70, 101-103).

Thus, the initial observations of Barker and colleagues, together with many subsequent

studies clearly indicate that a broad range of inflammatory diseases have their origins in

fetal life. It is also evident that events in utero and the early postnatal period are

important in determining the propensity for inflammation that contributes to the

subsequent disease risk. This relatively new field of medicine holds much promise for

explaining the rise of NCD. However, many key questions remain. To what extent do

transient exposures really result in long-term adverse outcomes in the offspring? Which

exposures are important and what is the mechanism through which their effects are

exerted? This thesis aims to contribute to this area by examining effects of specific

nutritional exposures on early gene expression in humans using cutting edge epigenetic

profiling technologies.

1.3 Gene-environment interactions in determining disease risk

Despite the extensive body of data linking modern lifestyles to chronic inflammatory

diseases, not all individuals living in modern environments acquire these conditions.

One proposed explanation for this suggests genetic factors are important for

determining an individual’s susceptibility profile. The ‘gene-environment’ interaction

paradigm is a longstanding model through which to understand complex diseases. This

paradigm supposes “a different effect of an environmental exposure on disease risk in

persons of different genotypes” (104). Following the completion of the human genome

project in 2003, a decade of genome-wide association studies (GWAS) studies sought to

identify disease-causing variants that might serve as predictive markers or preventive

targets. However, despite a large investment in human population genetics, for most

complex diseases, these endeavours have failed to attribute any substantive proportion

11

of risk to genetics, or consistent gene-environment interactions (105-107). Moreover, a

genetic-based model cannot sufficiently account for the rapid cumulative rise in many

immune-related diseases since, in evolutionary terms, emergence of genetic traits

conferring disease risk may take many generations, much more than the documented

time frame for escalating rise in NCD. Therefore, the focus has been in identifying

mechanisms that have the potential to regulate expression of genes, independently of the

primary DNA sequence, that interact with the changing environment.

The emerging field of epigenetics provides a new frontier for understanding how

environment influences the expression of genes. Unlike DNA marks, epigenetic marks

encode information from both the inherited genotype and environmental exposures and

thus represent a promising approach to explain pathogenesis of complex diseases.

1.4 Epigenetics: what is it?

Epigenetic processes are key determinants of gene expression, and as such they now

appear to be the key mediators of gene-environment interactions, providing new

dimensions of interest in epigenetic mechanisms of disease risk.

Epigenetics can be broadly defined as a series of complex, dynamic and interconnected

biological processes that regulate the expression of genes, to produce changes in cellular

function from a constant and unchanging underlying DNA sequence (108). This allows

the extensive cellular diversity within organisms and allows the functional flexibility in

response to changes in internal and external conditions. New insights into the

mechanisms of how gene expression can be altered by a range of environmental factors

has become the cornerstone of the DOHaD paradigm (109). These epigenetic processes

include DNA methylation, post-translational modification to histone tails and chromatin

remodelling factors and non-coding RNAs (ncRNA). Modification of gene expression

through these epigenetic processes controls a vast array of biological functions such as

cell differentiation, genomic imprinting and aging. Furthermore, information stored in

the epigenetic “code” can be propagated through mitosis and meiosis (110). Thus,

epigenetic information is not only maintained in daughter cells throughout life, but also

has the potential to be passed on to subsequent generations. The latter, also known as

epigenetic inheritance has been demonstrated in plants, in which a defined methylation

pattern has shown to propagate for many generations (111) and also in animals (112)

however, the extent and mechanisms of such inheritance in humans is yet not clear.

12

1.4.1 Molecular mechanisms of epigenetics

Eukaryotic genomes are packaged into chromatin, whose basic repeating unit is a

nucleosome that consists of a histone octamer wrapped around 147 base pairs (bp) of

DNA (Figure 1.1). The functional outcome of the genetic code largely depends on the

accessibility of the DNA to transcriptional machinery. Epigenetics provide a mechanism

by which the access to DNA is regulated resulting in either transcriptional activation or

repression of the gene. Through these mechanisms, over 200 cell types that comprise

the human body can be specified from a single genetic code. The epigenetic processes

that modulate the access to DNA in response to upstream signals include DNA

methylation, covalent modification of histones and chromatin interaction with

regulatory ncRNA and are described in detail in the following sections.

Figure 1.1 Schematic representation of the organisation and packaging of

nucleosome.

1.4.2 DNA methylation

DNA methylation in mammals, the most widely studied epigenetic process, consists of

transfer of a methyl moiety from S-adenosylmethionine (SAM) to the 5-position of the

cytosine residues in certain CpG (cytosine followed by a guanine, linked together by a

phosphate) dinucleotides. This transfer reaction is catalysed by DNA

methyltransferases (DNMT) that include family members DNMT1, DNMT2 and

DNMT3. The addition of methyl groups to DNA generally induces transcriptional

silencing by blocking the binding of transcription factors and/ or through promoting the

binding of methyl CpG-binding domain (MBD) proteins, which recognise 5-methyl

cytosine (5-mC) on DNA. Such proteins bind to 5-mC, which in turn recruits histone-

modifying complexes to the site resulting in a closed chromatin structure and

transcriptional silencing (113). Recently, Severin et al reported that DNA methylation

may also regulate gene expression through altering mechanical properties of DNA

13

including flexibility of DNA which is important for DNA to wrap around histones,

hence accessibility of DNA (114).

CpG dinucleotides are unevenly distributed throughout the genome. In general, CpG are

under-represented in mammalian genome due to evolutionary spontaneous deamination

of 5-mC to thymine (115). However, certain regions of DNA contain a high density of

CpG and are referred to CpG islands. They often span sites of transcriptional initiation.

Typically 60-70% of annotated gene promoters are associated with a CpG island,

including most house-keeping genes. These CpG islands are mostly unmethylated

favouring a constant expression of these important genes. In contrast, the majority of

non-CpG island sites are methylated, such as repetitive sequences and mobile elements

ensuring genome stability through silencing these transposable elements (116).

However, new data suggest that methylation is not exclusive to CpG sites as previously

thought, but also occurs at non CpG sites such as CpA (cytosine followed by an

adenosine) and CpT (cytosine followed by thymine) (117, 118).

DNMT1 family is important for establishment and regulation of tissue-specific patterns

of methylated cytosine residues. This enzyme copies the pre-existing methyl marks on

to the daughter strands during DNA replication to maintain the established methylation

patterns across successive cell divisions and given the name “maintenance

methyltransferases” since they add a methyl group to DNA of the unmethylated strand

when the other strand is already methylated (hemimethylated) (119). DNMT3 family is

different to DNMT1 such that it is mainly responsible for de novo methylation and is

abundantly expressed in early embryo. This family of DNMT include, DNMT3A and

DNMT3B and one regulatory factor DNMT3-like protein (DNMT3L), which has some

structural similarities and differences to the rest of DNMT3 proteins. DNMT3 is

important during embryogenesis to establish de novo methylation marks in the zygote

that were lost globally shortly after fertilisation (120).

DNA methylation is a relatively stable epigenetic mark established primarily during cell

differentiation. Each cell type in the mammals contains a unique DNA methylation

signature that confers cell-specific functions. Despite these being very long-lived

modifications, DNA methylation is also dynamic and responsive to environmental

factors, conferring phenotypic plasticity among related cell types (121). Removal of

methyl groups (DNA demethylation) is an active process mediated by ten-eleven

14

translocation proteins (TET) proteins that remove or hydrolyse the methyl group from

5-mC (122). Also, passive DNA demethylation has been observed during successive

rounds of replication in the absence of functional DNA methylation maintenance

machinery. Animal experiments have provided evidence that 5-hemimethyl cytosine (5-

hmC) may not merely be an intermediate in the demethylation pathway but may have a

distinct epigenetic role (123). However, the exact mechanism and functional

consequences of TET-mediated DNA demethylation remain to be clarified.

The work of this thesis involves genome-wide profiling of DNA methylation patterns in

neonatal immune cells to identify areas of methylome that are sensitive to early

environmental cues, and that may modulate immune development.

1.4.3 Histone modifications

Chromatin is a dynamic structure that responds to upstream signals to regulate access to

DNA. DNA is organised into nucleosome subunits of packaged DNA via histone

proteins. The nucleosome core consists of DNA wound around an octamer of 2 copies

each of the core histone proteins H2A, H2B, H3 and H4 (Figure 1.1). Unstructured tail

domains of histones are the sites where modifications take place. These modifications

represent major pathways in gene transcription, replication and repair. The structure of

chromatin can be altered in several ways including, covalent modifications of histone

tails, replacement of core histones with specialized variants (124) and repositioning or

eviction of histones from DNA by ATP-dependent chromatin remodelling enzymes

(125). Of these, modifications to histone tails has been extensively studied given the

fact that such modifications correlate well with the transcriptional activity of genes and

a large number of modifications have been described to date (126).

Acetylation of histone lysine tail residues carried out by histone acetyltransferases

(HAT) facilitates gene transcription. To describe how acetylation could favour gene

expression, it has been proposed that acetylation neutralises the positive charge of lysine

residues, weakening charge-dependent interactions between a histone and nucleosomal

DNA, linker DNA and/or adjacent histones, thereby increasing the accessibility of DNA

to the transcription machinery (127). Recent data show, in addition to direct influence

on increased accessibility of DNA, histone acetylation could favour binding of protein

modules that regulate transcription and chromatin dynamics (128). Advent of such

histone binding proteins may provide opportunities to discover molecules/particles that

15

have capacity to interact with histones to modify the protein-histone interactions to

influence the gene expression.

Unlike acetylation of histone lysine residues that are associated with transcriptional

activation, lysine methylation can be associated with either transcriptional activation

(H3K4me3 and H3K36me3) or repression (H3K9me3 and H3K27me3). Histone

methylation inhibits recruitment of polycomb repressive complexes, a family of

polycomb group proteins that through their enzymatic activity initiate and maintain

transcriptional silencing. Interaction of polycomb repressive complexes with methylated

lysine residues renders chromatin refractory to transcriptional repression (129). In

contrast, histones that are associated with repressive methylation marks show increased

affinity to certain DNA binding proteins including polycomb repressive complexes that

promote gene silencing (130). Other covalent histone modifications including

phosphorylation, ribosylation, glycosylation and ubiquitylation have been described,

however, we still know relatively little about their specific role in relation to

transcription and their relevance to biological processes.

Analysis of acetylation status of histones gives invaluable information on epigenetic

regulation of gene expression. Comparison of acetylation marks between healthy and

disease status further enables discovery of causal pathways and targets for therapeutics.

This project also analyses the acetylation status of histones that are associated with

known-immune genes.

1.4.4 Regulation by non-coding RNA (ncRNA)

There is accumulating evidence that ncRNA such as microRNA (miRNA), small

interfering RNA, and long ncRNA represent an important class of newly recognised

regulatory molecules (131, 132). Of these, miRNA has been the interest for their

association, when dysregulated, with progression of human diseases. MiRNA are small

non-coding single stranded RNA molecules of 19-25 nucleotides in length and have

shown to control major cellular processes, including metabolism, apoptosis,

differentiation and development (133). Regulation by miRNA, often negative on target

genes, is achieved in one of two ways; miRNA can cleave target mRNA transcripts by a

miRNA-associated RNA-induced silencing complex (miRISC) or miRNA binding to

sites within the 3′ untranslated regions (UTR) of target mRNA, thus inhibiting the

initiation of translation via the miRISC (134, 135). Subsets of miRNAs are thought to

16

act as tumour suppressor genes or oncogenes, and their dysregulation is a common

feature of human cancers (136). There are some fascinating features of biogenesis and

regulation of miRNA. A cluster of miRNA may be controlled by one promoter, but one

miRNA may be encoded by multiple pre-miRNA. Also, one miRNA may target

multiple genes, and one gene may be targeted by multiple miRNAs (137, 138). Because

miRNA directly regulate protein expression without transcription, miRNA may respond

more rapidly than other epigenetic regulators that require gene transcription in addition

to protein translation. Epigenetic modulation by ncRNA is becoming increasingly

recognised as a significant processes in disease development, however, it does not fall

within the main scope of this thesis.

1.4.5 Chromatin remodelling complexes

A group of chromatin remodelling complexes have been described that physically alter

chromatin structure, thereby influencing the function of the chromatin (139). These

complexes contain a central ATPase component that harnesses ATP hydrolysis to

physically remove or slide histones from DNA. The chromatin-remodelling complexes

are categorized into four distinct groups: ISWI (Imitation SWItch), INO80/SWR1

(INOsitol requiring/ Sick With Rat8 ts), CHD (chromodomain helicase DNA binding

protein) and SWI/SNF (SWItch/Sucrose Non- Fermentable). ISWI complex, the best

characterized chromatin remodelling complex, has shown to affect the DNA repair,

DNA replication and transcriptional regulation (repression/activation) through

interactions with transcription factors, co-regulators or components of the transcription

machinery (139).

The list of epigenetic mechanisms being discovered continues to grow. However, DNA

methylation and covalent modifications of histones have so far been a central focus in

epigenetic research. While different epigenetic mechanisms follow different pathways

to regulate gene expression, they tend to be interrelated as part of a coordinated

regulation of chromatin dynamics.

1.5 Functional implications of epigenetic dynamics

The phenotypic outcome of a particular epigenetic process depends on the biological

pathway/pathways they are linked to. There is increasing evidence that epigenetic

regulation continues to regulate across the whole life span, from the first moments of

17

life until death. The following sections focus on epigenetic control involved in

development and immune maturation.

1.5.1 Epigenetic regulation during embryo development and haematopoietic

development

The erasure and re-establishment of DNA methylation patterns during mammalian

embryonic development demonstrates the dynamic nature of epigenetic regulation.

Remarkable changes to the epigenetic signature occur naturally during this period,

particularly when gametes fuse to form the zygote, and when gamete precursors, the

primordial germ cells (PGC) develop and migrate in the embryo (140).

Initially, in the zygote, the paternal and maternal genomes remain physically separated

and undergo different epigenetic reprogramming. The paternal genome is subjected to

both histone modifications and global DNA demethylation in the first cell of the zygote.

These changes are evident at a later stage in the maternal genome (in preimplantation

embryo) causing early asymmetry of the two epigenomes (141, 142). This epigenetic

asymmetry appears to be essential for normal embryonic development (143). Although

demethylation appears to be widespread across the genome, some genomic sequences

such as centromeric heterochromatin, imprinted loci and some retrotransposons escape

DNA demethylation during this preimplantation development (144, 145). These loci

being resistant to demethylation prevents activation of the silenced genes otherwise be

deleterious to the developing embryo. Sequence specific-DNA binding factors

cooperate with histone modifying machinery to protect these sites from loss of 5-mC.

Zfp57 and Trim 28, which are essential for maintaining methylation at imprinted loci

recruit DNMT and histone modifiers to maintain the methylation signature at imprinted

loci (146-148).

As the cleavage divisions progress, totipotency is lost and differentiation process

commences coinciding with the specification of the first cell lineages (149). This

process is largely accomplished by up-regulation of the de novo methylases, DNMT3a

and DNMT3b (150). The resulting new pattern of DNA methylation is then maintained

throughout subsequent cell divisions. The gene-specific epigenetic reprogramming is

essential to lock in gene expression programmes that specify cell fate. After

implantation, de novo methylation also ensures X-chromosome inactivation in female

18

embryos. Random X-chromosome inactivation involves chromosome-wide changes

silencing genes in the chosen allele (151, 152).

A second wave of epigenetic reprograming occurs at the stage of germ cell

development. The cells that are already specified to become PGC show first sign of

reprogramming during their migration to the genital ridges of the developing embryo

(153). Then they undergo extensive and rapid loss of 5-mC including demethylation of

the imprinted genes when they enter the gonads (154). In addition to DNA

demethylation, changes to histone proteins have also been documented. During their

migration PGC undergo a reciprocal loss of H3K9me2 and increase in H3K27me3

(155). Nevertheless, whether the histone modifications initiate the DNA demethylation

or these are two parallel epigenetic processes is not clearly known.

Through this process PGC genomes are ‘wiped clean’ of most epigenetic marks

associated with somatic gene regulation to acquire the pleuripotential capacity of the

germ cell-specific signature. During the reprogramming period PGC lose parent-of-

origin-specific differential methylation marks at imprinted loci and assume imprinting

marks appropriate to their own sex. While this erasure of epigenetic marks occur in a

similar time frame in males and females, reestablishment program diverge according to

sex. For example, in males, de novo methylation of germ cells in fetal testis is largely

completed at birth (156, 157). In contrast, in females, imprints in primary oocytes are

quiescent until they enter the growth phase in the postnatal period (158, 159).

Collectively, data points to the importance of epigenetic mechanisms in controlling

critical developmental processes during early phases of embryogenesis and disruption

of this regulation may have detrimental consequences for development.

Haematopoietic cell development is another key process governed by the epigenetic

machinery during fetal life. Epigenetic mechanisms regulate the development of

haematopoietic stem cells (HSC), myeloid and lymphoid progenitors (160, 161). Both

DNA methylation and histone modifications governed by polycomb repressor

complexes play a significant role in these processes (162, 163). There are other

examples of how epigenetic processes govern the fate of progenitors as they

differentiate into various cell types (164-166). Given the important role of epigenetic

mechanisms in governing the ordered process of hematopoietic development, there is

speculation that disruption to these processes may alter the function of key immune cell

19

types, and their resulting response capacity. In the present study we have focused on

leukocyte methylation profiles as they may harbour epigenetic variants relevant to

immune function.

1.5.2 Epigenetic profile of the neonatal immune system and epigenetic changes

during postnatal development

Epigenetic mechanisms play an important role in maintaining the unique immune

profile at the feto-maternal interface, supporting and facilitating successful pregnancy

through the distinct immune profile characterised by a Th2 dominant cytokine milieu. In

particular, hypomethylation of Fork head box protein 3 (FOXP3) in the placental tissue

(10) and specific DNA methylation profile of macrophages (167) are important for

maintaining an anti-inflammatory environment at the feto-maternal interface.

It is now also evident that epigenetic mechanisms play a crucial role in directing

neonatal responses, controlling the expression of key cytokine genes in Th cells towards

Th2-biased neonatal responses, relative to the more Th1-dominant responses of adults

to the same stimuli (24). CD4+ T-cells are central to these effector immune responses

and many epigenetic mechanisms converge to build a unique epigenetic profile in

neonatal CD4+ T-cells to meet the functional requirements of this critical developmental

stage (168-170). In addition to epigenetic differences in CD4+ T-cell compartment,

epigenetic changes also underlie the functional differences in innate immune cells such

as DC (171, 172) and monocytes (173). A previous study in my host laboratory

characterised the longitudinal changes in DNA methylation of mononuclear cells (MC),

revealing significant methylation changes in genes associated with immune pathways

from birth to 5 years of life (174). Collectively, these observations suggest that

epigenetic mechanisms make an important contribution in the regulation of immune

maturation.

Collectively, these studies provide an epigenetic basis for the significant reprogramming

of immune function required as the fetus transits from the uterine environment, where

immune tolerance is key to survival, to the extra uterine environmental where defence

against potential infectious threats becomes increasingly important.

20

1.5.3 Evidence of disruption of epigenetic pathways in immune diseases

An increasing number of studies show that epigenetic variations are associated with

many chronic diseases such as asthma and allergic diseases. Such association studies are

helpful to identify potential biomarkers or targets for therapy. Analysis of DNA

methylation profiles of patients with seasonal allergic rhinitis was able to clearly

distinguish patients from healthy controls indicating the potential of epigenetic analysis

to identify methylation marks as biomarkers in a clinical setting (175). Epigenetic marks

on disease-associated genes also vary with severity of the disease or the response to

treatment indicating the diversified benefits of epigenetic analysis in a broad spectrum

of clinical scenarios (176-178).

Genome-wide epigenetic profiling, when combined with pathway analysis, provides

information regarding potential biological pathways linked to disease pathogenesis. For

an example monocytes (179) and keratinocytes (180) from patients with atopic

dermatitis show aberrant DNA methylation patterns associated with IgE pathway and

TSLP pathway respectively. Further studies on atopic dermatitis revealed that miR-155

was up-regulated in skin lesions indicating disease pathogenesis involves disruption of

many complex and coordinated epigenetic processes (181). A recent study mapping

histone modifications in peripheral blood CD4+ T-cell subsets from asthmatic

individuals identified disease-specific enhancers and enhancers that are associated with

known single nucleotide polymorphisms (SNPs) related to asthma showed significant

changes in histone marks along the Th2 differentiation pathway (182). This is an

example of how epigenetic studies bring new insight into mechanisms of complex

diseases.

Genome-wide or candidate gene-specific case control studies have identified additional

loci that undergo differential epigenetic modifications and are summarised in Table 1.1.

These findings, together support that immune pathways are under epigenetic control and

development of disease parallels disruption of these epigenetic dynamics.

Human observational studies continue to provide a link between aberrant epigenetic

pathways and immune dysregulation, however, it is difficult to determine whether

epigenetic variation is the true cause or simply an effect of disease development in

human studies. Animal experimental work provides direct evidence that key pathways

in immune response are epigenetically regulated and that these are causally related to

21

Table 1.1 Gene loci that show epigenetic modulation in association with immune

diseases

Disease/ Phenotype

Tissue/ cell type

Gene loci associated with immune

disease/pathway

Associated epigenetic changes

Asthma Buccal cells ARG promoter DNA methylation inversely correlated with exhaled nitric oxide (183)

Asthma Treg cells and effector T-cells

FOXP3 and IFNG Increased DNA methylation in asthmatics (184)

Asthma Airway epithelial cells

STAT5A and CRIP1

Differential methylation in asthmatics (185)

Asthma Whole blood IL4R Methylation of cg09791102 site showed an association with asthma (186)

Asthma CBMC ASCL3 Methylation at 5’-CpG island of ASCL3 was associated with asthma (187)

Asthma Whole blood ADRB2 Higher methylation at 5’ UTR of ADRB2 was associated with asthma (188)

Asthma CD19+ B lymphocytes

CYP26A1 Differential methylation in house dust mite allergic asthma (189)

Systemic Lupus Erythrocytosis

CD4+ T-cells NRLP2, CD300LB and S1PR3

Differential DNA methylation profiles correlated with clinical phenotype (190)

disease pathogenesis. Mice deficient in Th2 locus controlling region in CD4+ T-cells

show a loss of general histone H3 acetylation and histone H3-K4 methylation, and

demethylation of DNA in the Th2 cytokine locus (191). In a mouse model of allergic

asthma this is associated with a marked reduction in eosinophil and lymphocyte

recruitment in the airways, decreased serum IgE levels and lung inflammation when

challenged with ovalbumin (191). Another murine study demonstrated that allergen

challenge is associated with an increase in DNA methylation at interferon gamma

(IFNG) promoter in CD4+ T-cells, with concomitant decrease in IFN-γ production

(192). This study also provided evidence that pharmacological treatment can reverse

these epigenetic changes. Others have shown that silencing of the IFNG gene may also

involve histone modifications at this locus (193). Specifically, in a mouse model of

Th2-driven allergic asthma, the chemical inhibition or loss of SUV39H1, a histone

methylase that trimethylates H3K9, skews T-cell response towards Th1 response and

22

decreases lung pathology (193). Several other animal experiments also show that

pathogenesis of allergic airway inflammation involves pathways regulated at the level

of miRNA (194-196).

Thus, epigenetics plays a significant role in regulating many important biological

processes. In particular, animal models provide convincing evidence that disruption of

epigenetically regulated pathways are causally related to immune diseases. In humans

there is also growing evidence that disease states are associated with underlying

dysregulation of functionally relevant epigenetic pathways. There is a recognised need

to understand how environmental risk factors may be driving these underlying

epigenetic changes to promote disease, as targets for intervention.

1.6 Epigenetics as an underlying mechanism of Developmental Origin of Chronic

Diseases

As described in section 1.2 a significant component of the risk of chronic disease is

conditioned during initial programming of physiological and metabolic responses to

environmental cues perceived during critical stages of early development (86, 87, 197).

Evidence both from human observational studies and animal experiments now suggest

epigenetics is the likely mediator of early gene-environment interactions that may

modify the susceptibility to NCD (89, 198). Furthermore, aberrant epigenetic regulation

during fetal period may be implicated in disease pathogenesis, as suggested by detection

of altered disease-associated epigenetic marks at birth (199, 200). Thus, factors that

influence the epigenetic processes during fetal development may alter gene expression

profiles resulting in phenotypic and functional changes that may predispose to disease

risk. Diet, smoking, exposure to microbes and exposure to endocrine disruptors and

other pollutants during pregnancy are some of the maternal factors that are associated

with modulation of the neonatal epigenome and are further discussed below. These

environmental culprits have also been associated with developing immune diseases

indicating the role of epigenetic regulation mediating the environment induced-disease

risk. Understanding the epigenetic effects of maternal exposures help to identify

epigenetic processes amenable to environmental modulation that might serve as

preventive or treatment targets.

23

1.7 Maternal environmental exposures that modulate the neonatal epigenome

The fetus is completely dependent on the mother, for all nutrients. Maternal health and

the maternal environment are of critical importance for all aspects of development, and

a broad range of associated factors have now been identified to affect the fetal

epigenome. As the major focus of the thesis, maternal nutritional factors that modulate

offspring epigenome are discussed in detail in section 1.7.1 and an outline for other

factors known to affect the neonatal epigenome is given in section 1.7.2.

1.7.1 Nutritional exposures in early life and the fetal epigenome

Nutrition is arguably the most influential environmental factor during fetal

development. Prenatal nutrition influences fetal growth and the development of

physiological functions of all organ systems (92, 201-205). Similarly, postnatal

nutritional exposures are critical for the ongoing developmental maturation of many

organ systems and for optimal physiological functions (203, 206).

While it has been recognised for some time that nutritional changes could modify

patterns of gene expression with lasting influences on phenotype and function, it has

been only relatively recently identified that epigenetic mechanisms play a key role in

this process. External environmental pressures such as dietary exposures can lead to

subtle variations in epigenetic regulation of immune gene expression, which can

potentially lead to more profound effects on subsequent immune function, clinical

phenotype and disease risk. There are now good examples in animal models (93, 207)

of how dietary changes can directly influence the epigenetic machinery by inhibiting the

enzymes that catalyse DNA methylation or histone modifications or by altering the

availability of substrates necessary for enzymatic reactions. These alterations in

epigenetic marks lead to either enhanced or supressed gene expression with an altered

phenotype depending on the nature of the affected biological pathways (108). Mathers

has described the sequence of events that link nutritional exposures to later health

outcomes through epigenomic modifications, by the concept of four ‘Rs’: nutritional

factors are Received and Recorded by the epigenome, evidence of these exposures are

Remembered across successive cell generations and the consequences of these

exposures are Revealed as altered gene expression, cell function and ultimately, health

(208). This concept reiterates that fetal epigenome is sensitive to nutritional exposures,

thus can be modulated leading to an altered phenotype.

24

Using a mouse model, Li and colleagues showed that early postnatal over-nutrition

leads to a reduction in spontaneous physical activity in female mice that sustained into

adulthood (209). These changes were associated with extensive changes in

hypothalamic DNA that was also persisted into adulthood, providing potential

mechanistic basis for persisted physiological effects. Similarly, prenatal exposure to

famine (under-nutrition) has also been associated with persistent epigenetic differences

in humans. Individuals who were prenatally exposed to Dutch Hunger Winter in 1944-

45 had, 6 decades later, less DNA methylation of the imprinted insulin-like growth

factor-2 (IGF2) gene compared to their unexposed, same-sex siblings (210). A growing

number of studies have reported that epigenetic marks serve as a memory of exposure in

early life to inadequate or inappropriate nutritional factors as reviewed in (211) and

(212). These exposures may modify a range of epigenetic marks including, DNA

methylation, post-translational histone modifications, chromatin remodelling complexes

and miRNA.

As noted in section 1.1.4, changing patterns of maternal diet also has a major impact on

changing microbial diversity, specifically gut microbiome with effects on both maternal

immune and metabolic homeostasis with consequences for fetal immune and metabolic

development. It is intriguing to speculate that developmental influences associated with

altered gut microbiome may be induced by effects on host epigenetic machinery. This is

based on the observations in animal models that gut microbiota can directly modulate

host immune gene expression through epigenetic changes. (213-215). The murine

experiments show that microbial short chain fatty acid fermentation products regulate

host Treg development through modulation of histones (216, 217). Collectively these

studies suggest that changes in the gut microbiome modulate immune development

potentially through epigenetic effects.

Identifying individual components of the diet which exert epigenetic effects,

importantly affecting fetal developmental programing, is a huge challenge. This thesis

examines two important maternal dietary factors namely folate and n-3 PUFA which

potentially affect developmental programing through epigenetic mechanisms.

1.7.1.1 Folate and other nutrients involved in one-carbon metabolism

As a major focus of this thesis, dietary components such as folate, methionine and

choline are of much interest because of their role in DNA methylation through 1-carbon

25

metabolism (218). Folate is a B vitamin (vitamin B9), and it is needed both to

synthesize DNA – 3 out of the 4 nucleotides needed to synthesize DNA require a one-

carbon that is carried by folate – and the methyl groups that are used to regulate gene

expression that converts cytosine to methylcytosine (also come out of one-carbon

metabolism, or folate metabolism) (Figure 1.2).

Consequently, folate metabolism is intimately tied to the genome by affecting both

genome stability as well as epigenetic marks on DNA. Dietary folate, through a series

of reactions, is processed into metabolic co-factors that carry chemically activated

single carbons, referred to as ‘one-carbon’. In this metabolism, a carbon unit from serine

or glycine is transferred to folate-derived co-factors.. Other nutrients and metabolites

that can provide one-carbon metabolites include choline and its degradation products

betaine, dimethylglycine and sarcosine. Other co-factors such as niacin (vitamin B3),

riboflavin (vitamin B2), vitamin B6 and cobolamin (vitamin B12) are also required to

complete the oxidation cycle of one-carbon to either synthesize nucleotides or to

generate methyl groups. In eukaryotes, folate metabolism is compartmentalized in the

cytoplasm, mitochondria and nucleus (219). While one-carbon metabolism in the

cytoplasm is required for the synthesis of purines and thymidylate and the remethylation

of homocysteine to methionine, mitochondrial events are much complex, with varying

metabolic outcomes depending on the tissue type and the development stage.

Folate is readily available in fresh fruits and vegetables, but destroyed by cooking.

Deficiency of folate is associated with intrauterine growth retardation, preterm or fetal

death, Down syndrome, and neural cleft-related birth defects including neural tube

defects and oro-facial clefting (220-222). Based on evidence that improving folate

status from preconception lowers the risk of neural tube defects, daily folic acid

supplementation 400µg has been recommended for all women planning for pregnancy

(223, 224). Identification of the protective effects of periconceptional folic acid

supplementation against neural tube defects in neonates and the high rates of unintended

pregnancy led to the mandated fortification of flour and other grain products with folic

acid first in the United States and then in several other countries (225). This initiation

has been a success in preventing occurrence of neural tube defects, however, total folate

intake exceeding the recommended limit has been reported among women of child-

bearing age (226, 227).

26

Figure 1.2 Schematic illustration of one carbon metabolism. The diet contains folate monoglutamates and polyglutamates, which need to be hydrolysed to monoglutamtes by glutamate carboxypeptidase II (GCPII). Monoglutamates are subsequently absorbed through the apical membrane of the intestinal epithelium. Folic acid, a synthetic form of folate is more readily absorbed than folate. The monoglutamate form of 5-Me THF is the predominant folate in the circulation and stored in liver until redistributed to other tissues. Uptake of folate by peripheral tissues occur via Reduced Folate Carrier (RFC) and once enter the cell, 5-Me THF acts as a carrier of methyl groups unit in the folate-methionine cycle. Subsequently the methyl groups are utilised to synthesize S-adenosylmethionine (SAM), the universal methyl donor in all tissues.

Previous studies by our group have shown that many women continue to consume

higher than necessary doses of folic acid later in pregnancy, well beyond the window of

risk for neural tube defects (228). There is little evidence of any significant beneficial

effects of folic acid supplementation after the first trimester (229), and some studies

suggest that this may be associated with an increased risk of allergic outcomes

(discussed further below).

There has also been a specific interest in effects of maternal folic acid supplementation

on the immune development of the offspring. This gathered momentum following the

seminal study, by Hollingsworth et al. who demonstrated that maternal folate

27

supplementation induced an allergic phenotype, with associated epigenetic changes, in

the offspring (93). This folate-rich maternal diet induced methylation changes in 82-

gene loci in the offspring, resulting in increased airway hyperesponsiveness, airway

eosinophilia, and production of inflammatory cytokines. This trait was then inherited to

the subsequent F2 generation. Among these genes, RUNX3, a gene known to regulate T-

lymphocyte development and to suppress airways inflammation (230) was

hypomethylated with concordant transcriptional silencing of this gene in the progeny

(93). The effects of nutritional impact on F1 and F2 generation is largely considered as

“parental effects” as the developing embryo and its germline are simultaneously

exposed to environmental signal that may also influence the F1 and F2 epigenomes. The

“epigenetic memory” retained in F1 and F2 generations may translate to phenotypic

variation in later life (231). This is different from true transgenerational epigenetic

inheritance that is seen from F3 generation and beyond who were not exposed to initial

environmental signals that triggered the epigenetic changes.

It is not clear how acquired epigenetic signature successfully pass through epigenetic

reprograming (erasure and re-establishment of epigenetic marks) in the developing

embryo and is difficult to address in human studies, however, elegant animal

experiments provide evidence for transgenerational epigenetic inheritance with

phenotypic consequences. In this regard agouti mouse model in which mice carrying the

viable yellow agouti metastable epiallele (Avy) causing variable expression of agouti

protein in isogenic individuals is an elegant animal model to study the epigenetic

inheritance (231). The variable coat colour of isogenic Avy mice correlates to

methylation status at Avy locus and using this murine model Morgan et al found that the

distribution of phenotypes among offspring is related to the phenotype of the dam that

was inherited through the female germline (231). Studies investigating this model

further observed that offspring born to dams fed a methyl-supplemented diet had brown

(pseudoagouti) rather than yellow coats in parallel with hypermethylation at Avy locus

(232, 233). Of most importance maternal nutritional change not only influence their

immediate offspring’s coat colour, but the effect was augmented throughout subsequent

five generations without a quantitative change in the exposure i.e. with the exact same

diet (231). Transgenerational amplification of environmental effects through changes in

the epigenome may have important health implications and provide a new hypothesis

that a stable environmental exposure could lead to a persistently increase in related

disorders such as allergic diseases.

28

Subsequent observational studies in humans have also linked folate intake with a higher

risk of allergic diseases (228, 234, 235) although these findings have not been entirely

consistent (236, 237). Several human studies reported an association of altered DNA

methylation in offspring born to mothers with folic acid supplementation supporting the

findings of the animal studies. Steegers-Theunissen et al (238) found a positive

correlation between periconceptional maternal folic acid supplementation and infant

DNA methylation of IGF2 differentially methylated region (DMR). While a lower

DNA methylation of H19 DMR has also been reported in association with higher

maternal folic acid intake during pregnancy (239), other studies have found no

association between either maternal folate intake or cord folate levels and DNA

methylation of IGF2 DMR (239, 240) or specific promoter regions of IGF2 (241) or

LINE1 (242, 243). Interestingly, Fryer et al (244) analysed the CpG dinucleotide

methylation in 12 cord blood samples using a genome-wide methylation profiling and

found an association between cord plasma homocysteine levels (inversely correlated

with serum folate) and methylation patterns, with 298 CpG sites showing significant

associations with plasma homocysteine level. Chang and colleagues investigated the

influence of maternal serum folate on DNA methylation in 18-28 weeks old fetuses

following termination of pregnancy for fetal anomalies (245). They found a positive

correlation between the levels of maternal serum folate and DNA methylation in brain

tissue but not in other tissues indicating that exposure to folic acid during pregnancy

may have tissue-specific effects in the developing offspring.

Studies investigating underlying epigenetic mechanisms of in utero folate exposure on

offspring effects have mostly focused on genomic regions surrounding imprinted genes

and only a few human studies have used a genome-wide approach in mapping DNA

methylation changes at birth in relation to variation in folate exposure. The folic acid

pathway plays a critical role in fetal development yet its effects on the epigenome and

gene expression is incompletely understood. This study investigates the genome-wide

effects of in utero folate exposure on DNA methylation in neonatal immune cells. It is a

timely need to understand the epigenetic effects of folate intake in pregnancy on

regulation of immune maturation which plays a central role in pathogenesis of NCD.

In addition to DNA methylation, histone modifications are another important epigenetic

modification that is associated with changes in nutritional exposures. Of histone

modifications, changes to histone acetylation is closely associated with variation in

29

dietary components (212).Therefore, examination of combinatorial effects of DNA

methylation and histone acetylation of nutritional manipulation will give a better

understanding of the epigenetic programming and subsequent health outcomes of such

interventions.

1.7.1.2 Polyunsaturated fatty acids (PUFA)

Dietary content of more “western” diet is changing and of immediate relevance to this

thesis, decreasing intake of n-3 PUFA has been implicated in rising propensity of

inflammatory diseases (2). Along with a reduction in food rich in n-3 PUFA such as

oily fish, increased intake of food high in n-6 PUFA has shifted the n-3/n-6 PUFA from

around 1:1 to 1:5-1:2 (246). This change in fatty acid balance has a profound effect on

immune response as a result of the relatively inflammatory properties of the n-6 PUFA

relative to the anti-inflammatory products of n-3 PUFA. To address the relative

deficiency of n-3 PUFA as a potential culprit of dysregulated immune development

efforts are being made to restore the PUFA balance through supplementing with fish oil

in pregnancy and early infancy. Previous studies from our laboratory have consistently

shown that n-3 PUFA supplementation in pregnancy (in the form of fish oil) reduce

oxidative stress and inflammation (52) , with effects on T cell activation (247) which

include reducing Th2 propensity in neonatal immune cells (52-54, 247) and have

supported the finding other groups (248, 249). While this dietary intervention has

shown to affect many biochemical and physiological properties of cells and organs

(250) the exact molecular mechanism by which maternal intake of n-3 PUFAs exert

their multi-system benefits in the offspring has not been clearly elucidated.

Emerging data from animal studies suggest maternal PUFA intake has epigenetic effects

in the offspring (251, 252) indicating offspring immunomodulatory effects of such

dietary interventions may have an epigenetic basis. This is a poorly investigated area

albeit the enormous potential of epigenetic studies to explore the immunomodulatory

pathways and molecules of PUFA supplementation.

In a recent study involving pregnant women supplemented with n-3 docosahexanoic

acid (DHA) or a placebo from the second trimester until delivery, Lee et al measured

the DNA methylation changes in genes associated with Th1, Th2, Th17, and Treg

development, as well as LINE1 repetitive elements of cord blood mononuclear cells

(253). Although there were no significant differences in DNA methylation pattern in

30

genes when comparing the treatment and the placebo groups, overall n-3 PUFA

supplementation did have effects on methylation in the subgroup of neonates whose

mothers smoked during pregnancy. Specifically, in this risk group, n-3 PUFA

supplementation was associated with differences in methylation levels of LINE1

repetitive element. Collectively, these data suggests maternal n-3 PUFA intake during

pregnancy may modify the fetal epigenome, and that this may depend on other

environmental exposures.

1.7.2 Other maternal exposures that affect the fetal epigenome

In addition to maternal nutritional exposures there are number of other maternal

environmental factors affecting fetal epigenome that have been described. Studies

employing genome-wide epigenetic analysis to investigate such effects have made a

significant contribution to discover areas in the fetal epigenome that are sensitive to

maternal exposures.

Evidence is accumulating for a clear association between maternal smoking to specific

changes in off spring DNA methylation, some of which were in line with the findings in

non-pregnant adult smokers. This has important health implications as exposure to

cigarette smoke in pregnancy has many adverse effects on the fetus, including effects on

lung function and asthma risk (254, 255). Smoking in the last trimester has been

associated with early onset of airway hyper-reactivity (likely asthma) by the age of 1

year (256). Moreover, both maternal and grand-maternal smoking during pregnancy are

associated with increased risk of childhood asthma, suggesting a persistent heritable

effect (257).

Human studies linking methylome profiles and maternal smoking have employed

available fetal derived tissues such as placenta, buccal cells or cord blood. Recent

several studies have linked exposure to smoking in pregnancy with DNA methylation

change in the Aryl hydrocarbon receptor repressor (AHRR) gene, a gene that is involved

in the detoxification of chemicals found in tobacco smoke. A study that involved

screening of 1062 newborn cord blood samples using the Infinium

HumanMethylation450 platform (HM450) found hypomethylation of the AHRR and

Growth factor independent 1 transcription repressor, and hypermethylation of

cytochrome P450–1A1 and myosin IG (258). These findings have been replicated in

other birth cohorts (258, 259). Interestingly, other recent studies in adults have also

31

identified hypomethylation in the same region of the AHRR gene in association with

smoking in many tissues or cell types (260-263). Furthermore, there is now evidence

that prenatal tobacco exposure not only have effects on epigenetic profile at birth, these

effects can also persist into infancy (259) or adolescence (264). In addition to the gene-

specific effects of prenatal tobacco exposure, there is emerging evidence of maternal

smoking could alter the fetal epigenome in a global scale (265). However, genetic

constitution appears to influence over the epigenetic-environmental interactions in this

context (265, 266).

Polycyclic aromatic hydrocarbons (PAH) are one of the most wide spread organic

pollutants and also a major component of particulate matter (PM) of urban aerosol.

They are found in oil, coal, and tar deposits and they are also formed by the incomplete

combustion of carbon-containing fuels such as wood, coal, diesel, fat, tobacco, and

incense. Grilled, smoked or barbecued meat also appears to contain high levels of PAH

(267, 268). In addition to its carcinogenic properties (269), it has been found not only to

impair functions of airway cells and smooth muscle cells but also diminish

responsiveness to standard therapy given to asthmatics (270) and lead to cognitive

defects and fetal growth impairment with in utero exposure (271, 272). A novel

epigenetic marker for PAH-associated asthma has been identified and cord blood

mononuclear cells (CBMC) of children born to mothers who had been exposed to

considerable level of PAH showed hypermethylation of the acyl-CoA synthetase long-

chian family member 3 promoter (187). Furthermore, the exposure level was highly

correlated with increased risk of asthma symptoms in the offspring before age 5 years.

A recent study reported hypermethylation of the IFNG promoter to be associated with

PAH levels in cord blood of offspring of the exposed mother (273).

The largest contributor of traffic-related PM is diesel exhaust particles (DEP). Exposure

of mice to inhaled DEP for 3 weeks can augment the production of IgE upon intranasal

administration of Aspergillus fumigates (274). Hypersensitisation occurred through

hypermethylation of IFNG promoter with concomitant hypomethylation of IL4

promoter in DNA from splenic CD4+ T-cells. The effects of PM could be mediated in

the airways through induction of oxidative stress. Treatment of A549 cells

(adenocarcinomic human alveolar basal epithelial cells) with either PM-10 or H2O2

increased the expression and release of IL-8 which increased the HAT activity, hence

remodelled the IL8 promoter region (275) suggesting PM exert their effects through

32

chromatin remodelling of the susceptible genes. Given the widespread exposure of

human population to environmental pollutants, both indoors and outdoors, it is

important to research the epigenetic effects of these exposures as a potential mechanism

underpinning the maternal transmission of adverse health outcomes to the next

generations.

As noted earlier (in sections 1.1.3 and 1.1.4) prenatal and postnatal microbial exposure

provides a primary signal for the maturation of a balanced immune system, and now

there is evidence that these effects are mediated through microbe-induced modulation of

the host epigenome. In support of this, offspring of mothers living in rural farming

environments have shown to carry a different epigenetic profile compared to those who

were born to mothers not living on a farm (276-279). In mice, maternal exposure to

different microbes during pregnancy led to attenuation of asthmatic symptoms in the

offspring due to increased IFN-γ production accompanied by hyperacetylation at the

IFNG promoter (280). These findings together, are highly suggestive of the fact that

microbial exposure during pregnancy can modify fetal immune responses through

epigenetic mechanisms.

* * *

The epigenetic profile at birth reflects the accumulated changes over fetal life in

response to in utero exposures. Over the course of pregnancy many exposures can affect

the epigenetic profile to varying degrees to influence phenotype, resilience or

vulnerability to disease. This identifies the need to more fully investigate the effects and

associations of individual environmental exposures and epigenetic marks at birth, and

beyond. In this context, well-controlled randomised trials provide an ideal opportunity

to examine specific factors while minimising the effects of other confounding factors.

Ultimately, it is also recognised that none of these exposures varies in isolation and that

an understanding of the composite effects of early environmental interactions will be

needed. However a component approach is an important part of these initial

investigations. To that end, the studies of this thesis focus on the effects of key dietary

exposures, namely folate and n-3 PUFA, including analysis of samples collected in the

course of randomised controlled trials.

33

1.8 Challenges of epigenome-wide association studies

Epigenome-wide association studies (EWAS) have recently been enabled following

developments in new technology including microarray technology and next-generation

sequencing. These platforms offer unprecedented opportunities to study the epigenetic

effects on a genome scale. EWAS designs have become popular approaches to identify

potential epigenetic mechanisms in an unbiased fashion, with a view to hypothesis

generation. Despite their recent popularity, EWAS study designs are unique and contain

a number of unique challenges.

Given the fact that epigenetic marks are tissue specific it is logical to examine

appropriate tissues relevant to the disease of interest, however, this is not always

possible due to limited access to tissue samples in humans. It is even more difficult to

explore tissue-specific epigenetic aberrations in fetal programming and epigenetic

analysis may have to be carried out on readily accessible fetal tissues such as cord blood

and placental tissues collected at birth. On the other hand, cellular heterogeneity of

blood and other tissues is a potential confounder for epigenetic analyses. A number of

algorithms have now been developed to account for the inter-individual variation in the

composition of whole blood during data analysis (281). Use of purified cell populations

is a better alternative to overcome this problem in epigenetic studies.

Conclusively identifying causal from consequential epigenetic variants is a considerable

issue in EWAS as epigenetic variation can be causal for disease or can arise as a

consequence of disease (reverse causation). While some disease-associated epigenetic

variations have been detected prior to disease onset (282, 283) this is generally not

achievable for most phenotype-based retrospective studies and large longitudinal

cohorts provide an approach to rule out reverse causation of epigenetic variation. The

presence of highly correlated DNA methylation patterns across multiple tissue or cell

types, which otherwise have a unique epigenetic profile, in association with a disease

also suggest a causative link (284). Therefore, it is worth considering screening two or

more cell or tissue types for epigenomic analysis to gather supportive evidence for

causation.

Another aspect to consider in EWAS design is measuring heritable contribution to the

epigenetic variation (285). Underlying DNA sequence, at least for some loci is

important in determining the epigenetic status and therefore may influence how the fetal

34

epigenome respond to environmental exposures. Integration of genomic, epigenomic

and environmental data is a useful strategy to uncover the effects of genotype on the

epitype in an altered environment. Technology is now available to determine the precise

nucleotide positioning, genome-wide spread of methylated cytosines and histone

modifications across the genome enabling integration of multitude of information

during the analysis, but these strategies are likely to require large sample sizes to

achieve adequate power (286, 287). On the other hand, randomised controlled trials

offer unique opportunity to control the genetic confounding of epigenetic effects

induced by specific exposures through analysing repeated epigenetic measurements

before and after the intervention.

Despite the issues surrounding designing the studies, EWAS holds promise to unravel

gene-environment interactions in disease pathogenesis. While the field of epigenetic

epidemiology is still in its infancy, EWAS is likely to evolve as information and

experience accumulate and human epigenetic studies are essential to advance our

understanding of DOHaD.

1.9 Concluding remarks

It is clear that developing immune system is most vulnerable for modulation by

environmental factors during fetal and early postnatal periods as it is a period with the

potential to program or ‘steer’ the developing immune system towards an altered

phenotype.

Although some inherent plasticity remains across the life course, the appearance of

disease phenotypes in the first years of life, namely infant allergic disease, indicates

early consequences of different trajectories of immune development (4). While genetic

predisposition is important, surging rates of disease indicate that changes in the early

environment are driving these modifiable effects on early immune function. Because

inflammation and immune dysregulation are predisposing elements of many other later-

onset NCD, understanding how early events modulate the developing immune system is

also relevant to understanding the dynamics of inflammation later in life (96), and the

multisystem consequences. In particular, understanding how common risk factors for

many NCD modulate gene expression in early life is a research priority, as this may

provide better strategies for more targeted and more effective disease prevention.

35

Maternal dietary changes, whether as a general part of lifestyle change or as a specific

intervention, has consistently shown to affect the immune outcomes in the neonate (41,

53, 54, 288, 289). Nevertheless, the underlying mechanism of environment-induced

immune modulation is far from clear and only recently epigenetics emerged as likely

mechanism that underpin such effects. The work of this thesis aims to measure

epigenetic variation associated with in utero dietary exposures, specifically exposure to

folate and n-3 PUFA, which are known to influence immune development. This will

firstly, identify epigenomic regions that are sensitive to these specific environmental

factors and thus susceptible to modulation, and secondly, it will add to the scientific

evidence needed to inform evidence-based recommendations on dietary practice and

other strategies to promote favourable early immune development, ultimately aimed at

reducing the burden of both early and later onset inflammatory NCD.

36

Chapter 2

Study Rationale, Study Design and Methods

2.1 Study rationale

Based on clear evidence that periconceptional folic acid intake significantly reduces the

occurrence of neural tube defects in neonates, folic acid in doses of 400 µg daily has

been recommended to women of child bearing age in many countries (223, 224).

Mandatory folic acid fortification of flour and grain products in many countries has

been a public health success story to date, resulting in a significant reduction in

incidence of neural tube defects in newborns (290). Whilst these beneficial effects are

clearly warranted, post fortification studies have also raised some concerns over the

combined effects of public health recommendations and fortification of the food supply.

In particular there are legitimate concerns that pregnant women may potentially be

exposed to higher than recommended doses of daily folic acid (227, 291). Evidence

supporting these concerns was published in studies reporting a link between higher

folate intake during pregnancy, and an increased risk of asthma and allergic diseases in

the offspring. These data from observational studies have been inconsistent (228, 234-

237, 292), however mouse models have presented convincing mechanistic evidence for

such associations (93). Clearly more research is needed in this area in order to better

understand the role of folate in pregnancy and in the long-term health of the offspring.

The clear role of folate in one-carbon metabolism suggests a plausible mechanistic link

between excessive folate levels, aberrant DNA methylation at important immunity

genes, and potentially altered immune responses. In line with the DOHaD hypothesis,

these effects could potentially be programed through disrupting DNA methylation

levels during the establishment of the fetal epigenome. However, many studies in this

area have taken a candidate approach, limiting analysis for few target genes (238, 239),

and the full spectrum of genomic regions modulated by folate exposure are unclear.

This thesis investigates the effects of in utero folate exposure on DNA methylation

particularly in neonatal immune cells using an unbiased genome-wide approach. It also

investigates the effects of folate exposure on histone acetylation in the same population,

to understand the combinatorial effects of nutritional manipulation on different but

intimately related epigenetic processes.

37

In addition to the effects of folate there has been increasing interest in fish oil

supplementation during pregnancy and early infancy as a potential intervention to

restore the imbalance between relatively anti-inflammatory n-3 PUFA and more

inflammatory n-6 PUFA typically associated with modern diets. While n-3 PUFA have

been shown to exert multisystem effects promoting a healthy immune maturation, the

underlying mechanism is not clearly elucidated. Studies by Sadli et al (293) and

Ceccarelli et al (294) provided key evidence that fish oil may exert its effects through

DNA methylation. This thesis contributes to this emerging body of work, by

investigating the potential effects of maternal fish oil supplementation on the neonatal

epigenome, with a view to identifying previously unknown pathways in the

inflammatory cascade.

2.2 Study cohorts

The cohorts of this study were derived from participants from two previous research

projects carried out in our laboratory. The participants were recruited through Princess

Margaret Hospital for Children’s clinical research facility. An outline of the two studies

is given below.

Allergy Prevention Program (APP)

This double-blind placebo control trial was performed by Dr. Janet Dunstan (under the

supervision of Prof Susan Prescott) during the period between 1999 and 2002 at the

School of Paediatrics and Child Health, University of Western Australia to investigate

the effects of fish oil supplementation in pregnancy on infant outcomes. Ninety eight

atopic pregnant women were recruited to the study and received either fish oil or

placebo from 20 weeks of gestation until delivery. A total of 70 cord blood samples

collected from neonates of this cohort were available as cryopreserved samples and

were used to investigate the effects of maternal fish oil supplementation during

pregnancy on neonatal epigenetic profile as described in chapter 5.

Infant Fish Oil Study (IFOS)

This postnatal double-blind randomized clinical trial was performed by Dr. Nina D’vaz

and Dr. Suzanne Meldrum (also under the supervision of Prof Susan Prescott) during

the period between February 2008 and January 2011 at the School of Paediatrics and

Child Health, University of Western Australia. This study included 420 infants at high

38

risk of allergic disease (born to atopic mothers) who were supplemented with either fish

oil or placebo oil daily from birth to six months of age to determine whether infant fish

oil supplementation in the early postnatal period can favourably modify infant immune

development away from the allergic phenotype.

As this population did not have any interventions in pregnancy, it was the suitable

population to examine the effects of variations in maternal folate status in pregnancy, in

relation to the cord blood epigenetic marks. To investigate this, subgroups of neonates

with either high or low folate levels were selected from this cohort (based on the criteria

described in chapter 3). Since these neonates were supplemented with fish oil or placebo

in the postnatal period, the intervention did not influence on epigenetic marks at birth.

2.3 Data and sample collection

An overview of the available clinical and questionnaire data of aforementioned two

cohorts as well as available relevant samples is provided below.

2.3.1 Questionnaire data

Questionnaires were given to the mothers at their antenatal visits to collect data on

factors that may affect offspring immune development including familial allergy,

ethnicity, parental ages, number and age of siblings, socioeconomic and educational

status, use of medications, illnesses, home environment and lifestyle. A detailed account

of supplement intake and diet during pregnancy was collected at the recruitment through

semi-quantitative food frequency questionnaires.

Data on neonatal birth parameters, delivery mode, drugs used during the delivery,

complications during or after the delivery, and congenital abnormalities were available

for both cohorts. Follow-up questionnaires had been administered at 12 months to

collect data on potential symptoms of infant allergy (including eczema, food allergy,

and recurrent wheezing), infections, medication use, vaccination, daycare attendance

and exposure to other children, pets and tobacco smoke, feeding practices, solids

introduction and any changes that may have occurred in the home environment.

2.3.2 Maternal and infant skin prick test (SPT)

Maternal and infant (at 12 months) allergy was assessed by skin prick test to a panel of

inhalant and food allergens using a standardized technique (295). Allergens tested in the

39

mothers included cockroach, house dust mite, mould, southern grasses, rye grasses, cat,

dog and feathers. The infants were tested to egg, peanut, milk, house dust mite, cat, rye

grass and southern grasses. Saline was included as a negative control and histamine as a

positive control. A wheal diameter of ≥2 mm was considered positive.

2.3.3 Clinical assessment for allergic diseases

Infants were assessed clinically at 12 months of age, and the findings were reviewed by

a paediatric allergist. A child was classified as having “allergic disease” if they had a

physician diagnosis of IgE-mediated food allergy, eczema or asthma (although the

limitations in diagnosing asthma at this age are recognised). IgE-mediated food allergy

was defined by a history of immediate symptoms within 1-2 hours following contact

and/or ingestion and a positive SPT to the implicated food. . The diagnosis of atopic

dermatitis in infants was based on a history of typical skin lesions. The severity of

eczema was assessed using the modified SCORAD index (296). A diagnosis of asthma

was based on a history of recurrent wheeze (>2 episodes of wheezing) that was

demonstrated to be responsive to bronchodilator medications, but the limitations are

recognised at this age.

2.3.4 Collection of cord and maternal peripheral blood

The umbilical cord was cleaned and swabbed with 70% ethanol to prevent

contamination and cord blood was then collected by venepuncture (19G needle) of

placental vessels, and placed into heparinised (500 µl of preservative-free heparin)

Roswell Park Memorial Institute (RPMI) (Invitrogen, California, USA) tissue culture

medium. Twenty millilitres of maternal blood was collected via venepuncture of the

cubital fossa vein, into an equivolume of heparinised (500 µl of preservative-free

heparin) RPMI tissue culture medium. Cord and maternal serum was also collected and

stored separately for subsequent analyses.

2.4 Laboratory methods

Detailed methods are included in relevant chapters. Information relating to the methods

which are not described in detail elsewhere is described below:

2.4.1 Harvesting and cryopreservation of mononuclear cells

For logistic reasons and to allow batched analysis, cryopreserved cells were used for in

vitro analyses. These methods have been established and validated in the host laboratory

40

(297). Mononuclear cells were isolated from cord and maternal peripheral blood and

cryopreserved within 4 hrs and 12 hrs of collection respectively, as outlined below.

Cord blood samples were separated into plasma, mononuclear cell and erythrocyte

fractions by gradient centrifugation. Blood/RPMI was layered over Lymphoprep

(NYcomed Pharmacia, Norway) and centrifuged at 500g for 30 minutes. CBMC were

isolated from the layer above the concentration gradient and washed once with RPMI

and then layered over Lymphoprep and centrifuged at 500g for 20 minutes. This

“double-layer” technique ensures almost complete removal of contaminating

erythrocytes and nucleated erythroid precursors. CBMC were isolated again and

washed twice with RPMI + 2% heat-inactivated fetal calf serum (HIFCS) (Australian

Biosearch, Australia). A sample of cells were stained with white cell counting fluid and

were enumerated using an Improved Neubauer haematocytometer (weber Scientific,

West Sussex, England) and diluted to 30-40 x 106 cells/ml RPMI+2% HIFCS and

placed on ice. For cryopreservation an equal volume of cooled 15% dimethyl

sulphoxide (DMSO) (BDH, Australia) in HIFCS was added drop wise. Heat

inactivation of FCS (56ºC for 60 minutes) was performed prior to use.

Samples were frozen in 1ml aliquots each containing 15-20 x 106 cells. The samples

were cooled to – 80 ºC at a rate of 1ºC/minute in a specialized freezing container

(Nalgene cryocontainer) containing iso-propyl alcohol (BDH, Australia). Samples were

then transferred to a liquid nitrogen tank for long-term storage.

2.4.2 Thawing of cryopreserved mononuclear cells

Vials of cryopreserved CBMC were thawed rapidly in a 37ºC water bath. Cells were

transferred to a 10ml polypropylene tube and cold RPMI supplemented with DNAseI

(5µg/ml) (Sigma, Australia) was added drop wise (at 1ml/min) until the tube was filled.

The tubes were incubated for 10 min at room temperature to digest gDNA from dead

cells. The cells were centrifuged at 500g for 5 min and then resuspended to 1 ml in

AIM-V serum-free tissue culture medium (Invitrogen, California, USA) supplemented

with 2-mercaptoethanol ) (2ME) (4 x 10-5M final concentration) (Sigma, Australia) or

RPMI supplemented with 10% non-heat-inactivated FCS for determination of in vitro

tissue culture immune responses as detailed below. A sample of cells was stained with

(trypan Blue) and cell number and viability were determined using an Improved

Neubauer haematocytometer.

41

2.4.3 Cord blood immune responses

Cell suspensions were diluted in AIM-V supplemented with 2ME (for adaptive immune

responses) or RPMI + 10% non-HIFCS (for innate immune responses) to a

concentration of 1 x 106 cells/ml and seeded into 96-well round-bottom tissue grade

polystyrene plates at a volume of 250 µl/well alone or with optimal stimulating doses of

allergens or innate stimulants. The samples were cultured in duplicate where adequate

numbers of cells were available. Optimal allergen concentrations have been previously

determined by the host laboratory. Cells were incubated for 24 hrs and 48 hrs at

physiological 37ºC, 5% CO2 for innate and adaptive studies respectively. At the end of

the incubation period, 200 µl of culture supernatant was collected into microfuge tubes

and frozen at -20ºC for multiplex analysis of cytokine production.

2.4.4 Purification of CD4+ T-cells

CD4+ T-cells were chosen for this study to investigate epigenetic perturbations as they

play a key role in immune responses and have been shown to be susceptible to in utero

perturbation from environmental exposures (298).

The flow chart (Figure 2.1) below outlines the cell purification steps that were used to

isolate cell subtypes and indicates where the collected data on these cell subtypes are

represented in this thesis. CD4+ T-cells were isolated from CBMC using a two-stage

isolation strategy with magnetic DYNAL beads (Invitrogen, Victoria, Australia). 5 ml

of thawed primary mononuclear cells in AIM-V + 2ME tissue culture medium at a

concentration of 1 x 106 cells/ml was transferred each to two 10ml polypropylene tubes.

The volumes of buffers for this procedure were determined based on the starting CBMC

as per instructions from the manufacturer. The tubes were filled with PCF buffer

(Appendix 1) and centrifuged at 500g for 5 min at 8ºC to pellet cells. Supernatants were

aspirated and pellets were resuspended in 160uL of PCFD buffer (Appendix 1) and

combined with 33.6 µl of pre-washed magnetic beads coated with a monoclonal

antibody specific for human CD8. Cells were incubated at 4ºC for 20 min on a circular

rotor. Cells were gently resuspended and placed on a DYNAL magnet for 2 min before

removing the CD8- supernatant fraction. This fraction was added directly to 50.4 µl of

pre-washed magnetic beads coated with a monoclonal antibody specific for human

CD4. An extra 200µl of PCFD buffer was added to cells and incubated for 20 min at 4

ºC on a circular rotor. Meanwhile 350uL of RLT lysis buffer (Qiagen) supplemented

with 2ME (10 µl/ml) was added directly to CD8+ T-cell fraction, vortexed and frozen at

42

-80 ºC. Bead-bound CD4+ cells were washed twice in 1ml of PCF buffer and 350 µl of

RLT lysis buffer supplemented with 2ME was added directly onto beads, vortexed and

frozen at -80 ºC.

Figure 2.1 Outline of purification steps used to isolate CD4+ T-cells and antigen presenting cells (APC) from cord blood mononuclear cells (CBMC) for the studies in this thesis.

2.4.5 Purification of antigen presenting cells

Antigen presenting cells (APC) are critical regulators of host first line of defence and

dendritic cells (DC) in particular play a key role in the generation of adaptive immunity.

DC epigenome has shown to be sensitive to environmental cues (299) and variation in

epigenetic marks in DC and monocytes has been associated with disease risk (179, 199,

300). For epigenetic studies in this thesis, DC and monocytes were obtained from cell

sorting as described below.

The CD4-CD8- supernatant obtained following magnetic bead cell depletion (section

2.4.4) was transferred to a fresh 2 ml screw-cap tube and placed on a DYNAL magnet

for 1 min to remove any residual beads in the cell suspension that may interfere with the

process of cell sorting. Cells were transferred to a 5 ml polystyrene round-bottom tube

and were washed once with FACS buffer (Appendix 1). Cells were resuspended in

CD4+T-cells

CD8+T-cells

CD4-CD8-T-cells

APC cells

Fluorescence activated cell sorting

Methylation

Acetylation

CBMC

Methylation

Epigenetic

study

Representation

in the thesis

Chapter 3

Chapter 5

Chapter 3

Chapter 4

Chapte 3

Magnetic bead separation

43

100µl of antibody mix (20µl of anti-CD19-FITC, 10µl of anti-CD3-PE, 10µl of anti-

HLADR-PerCP, 2.5µl of anti-CD11c-PECy7, 2.5µl of anti-CD14-APCCy7 and 55µl of

FACS buffer). Whole CBMC were stained for single- colour controls (either with the

monoclonal antibody or with the appropriated concentration of matched-isotype control)

and used for determining the appropriate gating strategy. All antibodies were obtained

from BD Biosciences, CA, USA. After 30 min incubation in the dark at 4C, the cells

were washed twice with FACS buffer, and resuspended in 300µl PBS+20%FCS and

immediately placed on ice until run on the cell sorter (FACSAria) on the same day.

Shown in Figure 2.2 are representative FACS plots illustrating the sorting strategy that

was adopted to collect the monocyte/DC cell subsets from the CD4-CD8- fraction of the

CBMC samples. Figure 2.2A illustrates the FSC/SSC characteristics of the CD4-CD8-

fraction of CBMC. Figure 2.2B shows that the supernatant obtained following magnetic

bead cell isolation is depleted for CD3+ cells. From the CD3/CD8/CD4 depleted cell

population CD14+ (monocytes) and CD11c+HLADR+ dendritic cells were collected

(Figures 2.2 C, D and E) and were immediately resuspended with 350 µl of RLT lysis

buffer supplemented with 2ME, vortexed and frozen at -80ºC. Purities of sorted samples

containing DC and monocytes were 99-100% CD3-, 97-100% CD19-, 97-100% CD11c+

and 95-100% HLADR+.

2.4.6 Purification of genomic DNA and total RNA

Genomic DNA (gDNA) and total RNA were extracted from the frozen CD4+ T-cells

and APC using DNA/RNA mini columns (Qiagen, Australia) according to the protocol

recommended by the manufacturer. gDNA and RNA yield and purity were assessed by

spectrophotometry.

2.4.7 Quantitative polymerase chain reaction (qPCR)

First strand synthesis of complementary DNA (cDNA)

First-strand cDNA was generated by reverse transcription of 500 ng of total RNA per

sample with random hexamers using SuperScript VILO (Invitrogen, CA, USA)

according to the manufacturer’s protocol. RNA was treated with Deoxyribonuclease I

(Invitrogen, CA, USA) to remove any contaminating DNA prior to reverse

transcription. Reverse transcription was performed on an Applied Biosystems

thermocycler (GeneAmp 7000). cDNA were diluted 1/5 in RNAse free water and stored

at -20ºC.

44

Figure 2.2 Outline of screening strategy for isolating antigen presenting cells (APC) using FACS cell sorting. (A) FSC/SSC gating on the CD4 and CD8 depleted live cell population (B) CD3-CD19- cell population was selected and further gated for (C) CD14+ and HLA-DR. We collected the CD14+ monocytes (shown in C and D) and pooled these with the CD11c+HLA-DR+ cells as illustrated in (E) from the CD14-cell gate as per (C).

45

Real time polymerase chain reaction (PCR)

Guaranteed primer assays were purchased from Invitrogen and the sequence

information is proprietary of that company. Relative quantitation of gene expression

was performed using pre-designed TaqMan assays (Applied Biosystems, Foster City,

CA, USA) on the ABI 7300 real-time PCR system in triplicate. All samples were

normalized to the house-keeping gene PPIA.

2.4.8 DNA methylation analysis

A total of 500ng of purified genomic DNA (gDNA) from each cell type was bisulphite

converted using the Methyl Xceed kit (Human Genomic Signatures, NSW, Australia).

This procedure converts unmethylated cytosine residues into uracil while not affecting

5-methylcytosine. Successful bisulphite conversion was evaluated for all samples using

an in-house bisulphite-specific PCR. Bisulphite DNA was sent to Australian Genome

Research Facility (Parkville, Melbourne, Australia) for labelling, staining and

hybridization to Illumina Human Methylation 450 arrays (Illumina). Raw iDAT files

were processed using the Minfi package from the bioconductor project

(http://www.bioconductor.org) in the R statistical environment (http://cran.r-

project.org/). The minfi package was used for array pre-processing using the stratified

quantile normalization method. Technical bias attributable to different probe chemistries

between Type 1 and Type II probes were adjusted in this procedure. Probes with a

detection P-value call >0.01 in 1 or more samples were removed. Probes on the X and

Y-chromosomes were removed to eliminate gender bias. Probes previously

demonstrated to potentially cross-hybridize non-specifically in the genome were

removed (301). Probes containing a polymorphic single nucleotide polymorphism

(SNP) at the single-base extension site with a minor allele frequency of <0.05 were

removed. The log2 ratio for methylated probe intensity to unmethylated probe intensity,

the M value, was subsequently derived and used for statistical inference. Beta values

were derived from intensities as defined by the ratio of methylated to unmethylated

probes given by B=M/(U/M*100) and were used to compliment M-value as a measure

of effect size. Cluster analysis was used in conjunction with the chip-wide medians of

the methylated and unmethylated channels to identify any outlying samples.

2.4.9 Validation of methylation array targets

Probes that were shown to be differentially methylated in the 450k array were then

tested by mass spectrometry to determine the validity of the array findings. Amplicons

46

for targets were designed using the Sequenom EpiDesigner software

(http://www.epidesigner.com/) and performed using the Sequenom EpiTYPER platform

in triplicate. EpiTYPER (Sequenom, San Diego, USA) is a tool for quantitative analysis

of methylated DNA based on bisulfite conversion. Through bisulfite treatment

unmethylated cytosine residues are de-aminated to uracil and transformed into thymine

during PCR amplification, whereas methylated cytosine residues still appear as

cytosines. Consequently, bisulfite treatment results in methylation dependent sequence

variations of C to T after PCR amplification. Amplification products were treated with

shrimp alkaline phosphatase to dephosphorylate unincorporated deoxyribonucleotide

triphosphates from PCR. Subsequently, in vitro transcription was performed followed

by RNase A specific cleavage to produce smaller fragments. Cleavage products were

prepared for analysis in the mass spectrometer according to manufacturer’s protocol.

MALDI-TOF MS (matrix-assisted laser desorption/ionization time-of-flight mass

spectrometry) detects the 16 Da mass difference between guanine and adenine residues

(resulting from C/T variations at the opposite strand) due to methylated and

unmethylated DNA templates). MALDI-TOF MS data are then processed using the

EpiTYPER software generating quantitative results for each cleavage product. Probes

showing a % mean methylation of less than 5 and all probes overlapping with SNP were

removed from the analysis.

2.4.10 Histone 3 and Histone 4 acetylation analysis

Chromatin immunoprecipitation (ChIP) assay for histone acetylation studies requires a

higher starting numbers of freshly isolated cells. This has limited the use of this assay

primarily to cell lines rather than primary cell analysis. Therefore an in-house assay of

histone acetylation was developed and validated by our collaborating research group at

Philips University Marburg, Germany (described in detail in chapter 4). Using this

assay, levels of H3 and H4 acetylation of isolated neonatal CD4+ T–cells were measured

and analysed against the variation in folate exposure as described in chapter 4.

2.5 Statistical analysis

Statistical analysis is outlined in detail in the relevant chapters.

2.6 Ethics approval

Ethical approval for this study was obtained from the ethics committee of the Princess

Margaret Hospital for Children, Perth, Western Australia.

47

Chapter 3

Paper 1

Genome-wide DNA methylation profiling identifies a folate-sensitive

region of differential methylation upstream of ZFP57 imprinting

regulator in humans

Manori Amarasekera,1 David Martino,2, 3, 4 Sarah Ashley,2, 5 Hani Harb,6 Dörthe

Kesper,6 Deborah Strickland,7 Richard Saffery,2, 3 and Susan L. Prescott,1

1School of Paediatrics and Child Health, University of Western Australia, Perth,

Australia 2Cancer and Disease Epigenetics Group, Murdoch Children’s Research Institute,

Melbourne, Australia 3 Department of Paediatrics, University of Melbourne, Melbourne, Australia 4Centre for Food and Allergy Research, Melbourne, Australia 5Institute for Medical Research, Monash University, Melbourne, Australia. 6Institute of Laboratory Medicine, Pathobiochemistry and Molecular Diagnostics,

Philipps University Marburg, Germany 7Telethon Institute for Child Health Research, Perth, Australia

All authors are members of inFLAME (International Inflammation Network) of the

Worldwide University Network.

48

3.1 Abstract

Folate intake during pregnancy may affect the regulation of DNA methylation during

fetal development. The genomic regions in the offspring that may be sensitive to folate

exposure during in utero development have not been characterized. Using genome-scale

profiling we investigated DNA methylation in two immune cell types (CD4+ & antigen

presenting cells, APCs) isolated from neonatal cord blood, selected on the basis of in

utero folate exposure. High (HF, n=11) and low folate (LF, n=12) groups were selected

from opposite extremes of maternal serum folate levels measured in the last trimester of

pregnancy. A comparison of these groups revealed differential methylation at seven

regions across the genome. By far, the biggest effect observed was hypomethylation of

a 923bp region 3kb upstream of the ZFP57 transcript, a regulator of DNA methylation

during development, observed in both cell types. Levels of H3/H4 acetylation at ZFP57

promoter and ZFP57 mRNA expression were higher in CD4+ cells in HF group relative

to the LF group. Hypomethylation at this region was replicated in an independent

sample set. This data suggests that exposure to folate has effects on the regulation of

DNA methylation during fetal development, and this may be important for health and

disease.

49

3.2 Introduction

DNA methylation is a key epigenetic mechanism controlling fetal growth and

development (1). Animal studies suggest that dietary intake of nutrients involved in

one-carbon metabolism such as folic acid during pregnancy can influence DNA

methylation in the developing offspring that may lead to an altered phenotype (2-4).

In humans, periconceptional supplementation with folic acid is routinely recommended

to prevent the occurrence of neural tube defects (5). However, emerging data has linked

folic acid intake during pregnancy with disease outcomes in the offspring, including

atopic dermatitis (6, 7) childhood wheeze (8) and asthma (9). Parallel investigations into

potential underlying epigenetic mechanisms have mostly focused on genomic regions

surrounding imprinted genes that regulate growth and development. Several studies

have now reported variations in DNA methylation at these regions in offspring exposed

to periconceptional folic acid. Steegers-Theunissen et al reported the average

methylation of the imprinted insulin-like growth factor 2 (IGF2) differentially

methylated region (DMR) was 4.5% higher in infants whose mothers reported

periconceptional intake of folic acid at a dose of 400µg/day compared with infants

whose mothers did not take folic acid (10). Another study reported that average

methylation at the imprinted H19 DMR in cord blood leukocytes was significantly

lower in neonates of mothers who took folic acid either before or during pregnancy

compared to neonates of mothers who reported no folic acid intake as ascertained from

self-administered questionnaires (11). Together, these findings suggest a complex

relationship between maternal folic acid intake and DNA methylation levels at

imprinted genes and variation in folate exposure during development may have effects

on neonatal growth and susceptibility to diseases in later life (12). Adding another layer

of complexity to the association between folate and methylation profile is that the

effects of folate exposure appear to be tissue specific. Chang and colleagues

investigated the influence of maternal serum folate on DNA methylation in 18-28 weeks

old fetuses following termination of pregnancy for fetal anomalies. They found a

positive correlation between the levels of DNA methylation and maternal serum folate

levels in brain tissue but not in other tissues (13) indicating that exposure to folic acid

during pregnancy may have tissue-specific effects in the developing offspring.

Very few human studies have used a genome-wide approach in mapping the DNA

methylation changes at birth that are associated with variation in folate exposure during

50

pregnancy (14, 15). The concern has therefore been such that changes in epigenetic

marks in fetal epigenome induced by variations in maternal folic acid intake may have

long-term health consequences in accordance with the Developmental Origin of Health

and Disease hypothesis (DOHaD) (16). To advance this area, we conducted a genome-

scale profiling of 450,000 CpG sites in two key neonatal immune cell types (CD4+ T-

cells and antigen presenting cells, APC) purified from cord blood. We chose to study

two cell types representing distinct hematopoietic lineages with well-defined roles in

regulating the host immune response to the environment (17). Importantly by using

purified cell populations in this study, we were able to avoid potential confounding

effects of cell heterogeneity that may affect the measured epigenetic changes (18).

Neonates for this study were selected from the extremes of the maternal serum folate

distribution curve from a Western Australian birth cohort. Using an explorative

hypothesis-free approach coupled with an extreme of exposure design we have

identified several folate-sensitive differentially methylated regions (fDMRs) in the

genome.

3.3 Materials and Methods

Selection of the study population

The study population included 23 neonates selected from a larger prospective birth

cohort of mother-infant pairs recruited through the allergy research clinic in the Princess

Margaret Hospital for Children (7). Mothers were recruited during the last trimester

(>=28 weeks) of pregnancy at which time maternal blood samples were collected, and

cord blood samples were subsequently collected at the time of birth. Peripheral blood

mononuclear cells were harvested from blood samples within 12 hours of collection

according to standard protocols (19). Matched serum folate measurements were

available for both maternal and cord blood for n=222 mother-infant pairs. Extensive

clinical and dietary data collected at recruitment through semi-quantitative food

frequency questionnaires and maternal antenatal and socio-demographic factors were

also available.

The sample population was selected based on the following criteria: Infants were ‘non-

atopic’ and otherwise healthy based on clinical assessments and skin prick test to a

range of inhalant and dietary allergens conducted at 12 months. The high (HF) and low

folate (LF) groups were defined according to the first and third quartiles from the

distribution of maternal serum folate levels in conventional extremes of exposure

51

design. Infants were excluded from the study if exposed to maternal smoking in

pregnancy, or had evidence of congenital birth defects. All study procedures were

carried out in accordance with full institutional ethics.

Serum folate measurement

Folate levels were measured in maternal serum from blood samples collected during the

last trimester of pregnancy, and neonatal serum folate was measured from cord blood

samples using the Immulite 2000 competitive immunoassay platform (Siemans Medical

Solutions Diagnostics, Flanders, NJ, USA).

Isolation of CD4+ T cells

CD4+ T cells were isolated from cord blood mononuclear cells (CBMC) using a two-

stage positive isolation strategy with magnetic DYNAL beads (Invitrogen, Victoria,

Australia). CBMC were incubated with CD8+ magnetic beads as per the recommended

protocol (Invitrogen, CA, USA), and the CD8- fraction was then incubated with CD4+

magnetic beads as per the recommended protocol. Routine purity tests were conducted

by flow cytometry using antibodies CD19-FITC, CD3-PE, CD8-PerCP, CD11c-PE

Cy7, CD4-APC and CD14-APC Cy7 (BD Biosciences, CA, USA) and appropriate

concentration matched isotype controls. CD4+ cell purities ranged from 89-96% pure. A

fraction of isolated CD4+ cells were frozen with 15% dimethyl sulphoxide in heat-

inactivated fetal calf serum and stored in liquid nitrogen until transported to Marburg,

Germany for chromatin immunoprecipitation (ChIP) analysis. The rest of the CD4+ T

cells were lysed with RLT buffer containing 2-mercaptoethanol (Qiagen Allprep kit,

Qiagen, Victoria, Australia)) and stored at -80 ºC until genomic DNA was extracted for

methylation analysis.

Isolation of APC cells

CD3-CD19-CD14+/- CD11c+ HLADR+ myeloid APCs were sorted from CD4+ and CD8+

depleted cell fraction using FACSAria instrument and FACSDiva software (BD

Biosciences, San Jose, CA, USA).

Nucleic acid extraction

Genomic DNA and mRNA were co-purified from CD4+ T and APC cells using Qiagen

Allprep kits according to manufacturer’s instructions.

52

DNA methylation analysis

A total of 500ng of purified genomic DNA from each cell type was bisulphite converted

using the Methyl Xceed kit from Human Genomic Signatures. Successful bisulphite

conversion was evaluated for all samples using an in-house bisulphite-specific PCR.

Bisulphite DNA was sent to Australian Genome Research Facility (Parkville,

Melbourne, Australia) for labelling, staining and hybridization to Illumina Human

Methylation 450 arrays (Illumina). Raw iDAT files were processed using the Minfi

package from the bioconductor project (http://www.bioconductor.org) in the R

statistical environment (http://cran.r-project.org/). The minfi package was used for array

pre-processing using the stratified quantile normalization method. Technical bias

attributable to different probe chemistries between Type 1 and Type II probes were

adjusted in this procedure. Probes with a detection P-value call >0.01 in 1 or more

samples were removed. Probes on the X and Y-chromosomes were removed to

eliminate gender bias. Probes previously demonstrated to potentially cross-hybridize

non-specifically in the genome were removed (20). Probes containing a polymorphic

SNP at the single-base extension site with a minor allele frequency of <0.05 were

removed. The log2 ratio for methylated probe intensity to unmethylated probe intensity,

the M value, was subsequently derived and used for statistical inference. Beta values

were derived from intensities as defined by the ratio of methylated to unmethylated

probes given by B=M/(U/M*100) and were used to compliment M-value as a measure

of effect size. Cluster analysis was used in conjunction with the chip-wide medians of

the methylated and unmethylated channels to identify any outlying samples, and 1

sample was removed from the data set.

Quantitative PCR

First-strand cDNA was generated by reverse transcription of 500 ng of total RNA per

sample with random hexamers using Superscript VILO (Invitrogen, CA, USA)

according to the manufacturer’s instructions. cDNA was diluted 1:5 in RNAse free

water for gene expression analysis. Relative quantitation of gene expression was

performed using pre-designed TaqMan assays (Applied Biosystems, Foster City, CA,

USA) for ZFP57 on the ABI 7300 real-time PCR system in triplicate. All samples were

normalized to the house-keeping gene PPIA. Absolute levels of ZFP57 mRNA

expression were low and not measurable for 8/21 individuals and these were assigned a

maximum CT value of 45 for statistical analysis and interpretation at the group level.

53

Data analysis

Characteristics of genome-wide DNA methylation were assessed by principal

components analysis in the presence of potential confounding factors. The first fifteen

principal components capturing over 65% of the total variance were derived and tested

for associations with clinical variables. Clinical variables associated with variation in

DNA methylation are provided in the supplement and were included as covariates in

regression modelling. To identify differentially methylated regions (DMRs) we used the

dmrFind algorithm in the charm package under the default settings with the cluster

marker function. This algorithm combines surrogate variable analysis (21) for

modelling unexplained variability due to batch effects or covariates, with regression

modelling fitted to the entire dataset in the presence of covariates and surrogate

variables (22). Comparisons are made at the regional level as opposed to single CpG

analysis whereby methylation estimates are smoothed over spatial blocks accounting for

correlation between nearby CpG. Ontology enrichment analysis was performed using

the GREAT ontology tool under the default basal plus extension settings. Significant

ontologies are reported as raw -log10 binomial P-value <0.05.

Validation of array targets

Target validation was performed using the Sequenom EpiTYPER platform in triplicate.

Amplicons were designed using the Sequenom EpiDesigner software

(http://www.epidesigner.com/). Amplification conditions were as follows: 95° C for 10

min, 95° C for 15 s, 56° C for 30 s, 72° C for 2 min for 5 cycles, 95° C for 10 s, 60° C

for 30 s, 72° C for 1 min 30 s for 30 cycles, 72° C for 7 min. The target sequences of the

chromosome 6 fDMR were; aggaagagagGAGGATTTTAGAGGTTGGAAGTTTT (left)

and cagtaatacgactcactatagggagaaggctCCCTCTCATCTAAATCAAAAAACAC (right ).

Histone 3 (H3) and Histone 4 (H4) acetylation analysis

Using a validated ChIP analysis for H3/H4 acetylation in human immune cells we

analysed the levels of H3/H4 acetylation at selected loci. In brief, DNA-histone

interactions in isolated CD4+ T cells were first cross-linked with 1% formaldehyde.

Subsequently, chromatin was sonicated to achieve the optimal DNA length for the

immunoprecipitation which was done using antibodies against acetylated H3 and H4

(Millipore, Darmstadt, Germany). IgG mock control was bought from Abcam, San

Francisco, CA, USA. Quantitative PCR was performed to measure the precipitated

DNA of investigated loci. Primers were designed for known transcripts of ZFP57 and

54

are as follows; ZFP57-001 distal (ZFP57_1D) cac cac cgg cta act ttt gt (forward) and cct

ggg caa aaa gag tga aa (reverse), ZFP57-002 proximal (ZFP57_2P) ccc agg ctg gtg ttg

tta ct (forward) and ggt ttg atg tgg ctt cct gt (reverse), ZFP57-002 distal (ZFP57_2D) cat

gga aga gat ctt aga gag tg (forward) and acc taa tgc aga gat gct aaa taa (reverse).

3.4 Results

Clinical characteristics of the cohort

Maternal serum folate levels ranged from 4.1 nmol/l to 172.0 nmol/l in the cohort from

which the study samples were derived, with first and third quartiles 25.6 nmol/l and

50.5 nmol/l respectively. The high (HF) and low folate (LF) groups were identified

from this cohort and defined according to the first and third quartiles from the

distribution of maternal serum folate levels in conventional extremes of exposure

design. The mean maternal serum folate measurement at recruitment was 74.6 nmol/l

and 16.8 nmol/l in the HF and LF group (P < .001), with corresponding cord blood

serum levels of 78.2 and 36.4 nmol/l (P < .001) respectively (Table 3.1). There were no

significant differences in the key neonatal parameters between HF and LF groups, with

the exception of folate intake during pregnancy (either as food, P = .02 or as

supplement, p = .006), and cord blood serum vitamin D levels (P = .04), which were

found to be significantly higher in the HF group (Table 3.1). Based on this, all

subsequent analysis was adjusted for any potential effects of vitamin D. There was a

significant linear relationship between maternal serum folate and cord blood folate

levels in the sample population (r = 0.75, P < .001) (Figure 3.1A).

Characteristics of genome-scale DNA methylation profiles

Genome-scale DNA methylation data were generated from two blood cell types

representing both the innate (APC) and the adaptive (CD4+) immune cell compartments.

These cell types play a well-defined role in antigen presentation and processing and

mediation of effector immune cell responses. Principal components analysis on Beta

methylation values was used to explore variation in neonatal epigenetic profiles. The

most substantial source of variation in the data set was attributable to cell type (APC or

CD4+). Multidimensional scaling (MDS) and hierarchical clustering of samples showed

widespread differences between cell types reflecting the alternative lineages of APC

(myeloid origin) and CD4+ cells (lymphoid origin) (Figure 3.2A). Differential analysis

of CD4+ and APC cells revealed a signature of 13, 424 CpG sites (approximately 3.5%

of the CpG sites measured) that differed by a minimum delta beta value of at least 20%

55

and reached genome-wide significance (5048 hypermethylated; 8376 hypomethylated

sites). Localization of these sites to functional regions in the genome revealed the

majority of these differentially methylated positions (DMPs) were localized to gene

bodies as opposed to promoters, and overlapped with flanking regions of CpG islands

(CpG island shore and shelves), or were found in open sea regions (not island-

associated) (Figure 3.2B). The DMPs were significantly enriched for genes involved in

leukocyte function and antigen mediated pathways (binomial P value <0.05) (data not

shown). Birth mode, maternal serum folate levels, folate status (high or low) and serum

vitamin D levels were included as covariates in data modelling.

Figure 3.1 Relationship between maternal serum folate and cord serum folate levels. (A) Samples in the discovery cohort and (B) samples in the replication cohort. The graph shows the correlation plot between maternal serum folate levels measured at the recruitment visit during the 3rd trimester and cord serum folate levels collected at birth.

56

Table 3.1 Population characteristics of the mother and infants in the discovery

cohort

Characteristic

High folate group (n=11)

Low folate group (n=12)

P value

Mothers at recruitment Age, (yrs [SE]) 31.9 (1.25) 32.1 (0.82) 0.95

History of allergic disease 8 (73%) 8 (67%) 0.75 Asthma 4 (36%) 3 (25%) 0.55 Hay fever 5 (45%) 7 (58%) 0.54 Eczema 5 (45%) 4 (33%) 0.55 IgE mediated food allergy 1 (9%) 1 (8%) 0.95 Sensitization 9 (82%) 10 (83%) 0.92 Had tertiary education 8 (73%) 11 (92%) 0.23 Not exposed to passive smoke 36% 33% 0.44 Infants

Male (%) 4 (36%) 5 (42%) 0.80 Gestation (wk [SE]) 39.7 (0.3) 39.2 (0.3) 0.28 Delivery method

Vaginal 6 (54%) 4 (40%) Caesarean section 5 (45%) 6 (60%) 0.51

Neonatal growth parameters Birth weight (g [SE]) 3420.3 (79) 3570.9 (128) 0.43

Length (cm [SE] 50.2 (0.5) 49.9 (0.9) 0.79 Head circumference (cm [SE]) 35.0 (0.3) 35.6 (0.5) 0.34 Serum folate

Maternal serum folate level at recruitment (nmol/l [SE]) 74.59 (6.1) 16.8 (1.6) <0.001* Cord serum folate (nmol/l [SE]) 78.15 (5.1) 36.4 (2.3) <0.001* Cord serum Vitamin D (nmol/ [SE]) 59.9 (7.8) 39.0 (5.7) 0.04* Maternal daily intake from supplements (from questionnaire data)

Folic acid (µg DFE/day [SE]) 730.9 (127) 236.6 (120.7) 0.006*

Maternal daily intake from diet (from semi-quantitative food frequency questionnaire)

Folate (µg DFE/day [SE]) 310.7 (36.4) 199.2 (33.7) 0.02* Vitamin D (µg [SE]) 3.8 (0.8) 4.4 (1.7) 0.56 Total Energy (kcal [SE]) 2271.9 (126) 2019.2 (294) 0.71 Protein (g [SE]) 88.9 (5.2) 83.7 (12.8) 0.71 Total fat (g [SE]) 88.8 (8.4) 79.4 (12.9) 0.37 Copper (mg [SE]) 2.2 (0.1) 1.8 (0.3) 0.22 Zinc (mg [SE]) 12.0 (0.8) 10.5 (1.3) 0.22 Retinol (µg [SE]) 512.3 (84) 538.4 (157) 0.56 Vitamin B6 (mg [SE]) 2.0 (0.2) 1.5 (0.3) 0.22 Vitamin B12 (mg [SE]) 3.4 (0.4) 3.7 (1.0) 0.96 Alcohol (g [SE]) 1.8 (0.8) 1.8 (1.1) 0.78 Coffee (g [SE]) 84.2 (58.8) 144.9 (69.2) 0.25 Vitamin A (µg [SE]) 1390.2 (106) 1215.4 (268) 0.5

Statistical comparisons by Chi square and Mann-Whitney U-tests. DFE, dietary folate

equivalent units.

* indicates significant P-values (P < 0.05)

57

Figure 3.2 Differential methylation analysis between CD4+ and APC cells. (A) MDS analysis of sample relationships based on whole genome DNA methylation profile (left panel). Hierarchical clustering of samples based on Euclidean distance (right panel). Samples are coloured according to cell type (green, APC; orange, CD4+). (B) Differential hypomethylation (left panel) and hypermethylation (right panel) according to genomic region. APC; Antigen presenting cells, MDS; Multidimensional scaling.

Differential methylation according to folate status

We first examined general features of DNA methylation between folate groups at

several regions in the genome including repeat elements, non-repeat CpGs and

imprinted genes. Folate status was not associated with changes in DNA methylation

levels at long interspersed nucleotide element-1 (LINE-1) repeat elements in the

genome of either cell type (Figure 3.3A). The average level of DNA methylation in the

genome as measured by the mean of all CpGs on the array did not differ according to

serum folate status in either cell type (Figure 3.3B). Similarly, average and total levels

of DNA methylation levels across CpG sites targeting known imprinted genes did not

vary according to folate status (Figure 3.3C). Collectively this data suggests the general

58

features of the DNA methylome in the offspring are similar regardless of folate

exposure during pregnancy.

Figure 3.3 Analysis of global patterns of DNA methylation between folate groups. (A) Sequenom EpiTyper analysis of LINE-1 DNA methylation levels. (B) Mean methylation level for all somatic probes on the Illumina methylation 450k array. (C) Mean methylation levels specifically for probes targeting imprinted genes on the Illumina methylation array. All boxplots show means with range.

Next the methylation values for all probes were regressed on folate status (HF or LF),

adjusting for known covariates including cell type, to identify fDMRs in the

methylome. We adopted a sliding windows approach which is more powerful than

single CpG analysis to identify broad DMRs consistent in both cell types by computing

smoothed estimates of regionally associated probes (22). Seven candidate regions,

defined as an average of 4 or more consecutively differentially methylated CpG sites,

were identified in this analysis (Table 3.2) including a 923 base pair region in

chromosome 6 of substantial hypomethylation in the HF group 3kb upstream of the

ZFP57 gene (Figure 3.4).

59

Table 3.2 Differentially methylated regions (DMR) according to the folate status

aHypomethylation, bHypermethylation

Figure 3.4 Folate sensitive differentially methylated region (fDMR) in chromosome 6. The figure shows smoothed Beta methylation values for LF (red) and HF (blue) groups, averaged for both cell types. The location and size of this DMR is shown in the genome browser track above the sliding window. The ideogram track shows the location of this region on chromosome 6. LF, Low; Low folate, HF; High folate, DMR; Differentially methylated region.

Validation of candidate loci and functional assessment of ZFP57

We measured DNA methylation levels in CD4+ from the initial discovery cohort of

samples interrogated by the DNA microarray using the Sequenom EpiTyper platform as

Table 3.2 Differentially methylated regions (DMRs) according to the folate status

DMR according to the folate status, irrespective of cell type

DMR Chr start end size (bp)

no. of probes

average DM

maximum DM

HF vs LF gene assocations function

1 6 29648161 29649084 923 19 0.19 0.28

hypo a

ZFP57 (-3692)

Maintenance of DNA methylation and genomic imprinting

2 17 37123638 37123949 311 8 -0.08 -0.12

hyper b

LASP1 (+97682)

Regulation of dynamic actin-based, cytoskeletal activities

3 1 76189707 76190008 301 5 -0.13 -0.23

hyper ACADM

(-185)

mitochondrial fatty acid beta-oxidation pathway

4 8 144120106 144120706 600 7 0.08 0.10 hypo

LY6E (+20504) Lymphocyte antigen complex

5 1 228075423 228075749 326 5 -0.10 -0.13 hyper WNT9A

(+60090) development and cell fate

6 21 47604052 47604654 602 5 0.09 0.11 hypo

C21orf56 (+20) unknown

7 2 202901045 202901470 425 5 -0.08 -0.11 hyper FZD7

(+1948) Wnt protein binding

60

a gold standard technology. We designed an amplicon spanning a 321 base pair region

capturing 8 CpG sites in the chromosome 6 DMR upstream of ZFP57 identified by

HM450 analysis as broadly hypomethylated in HF samples. Data from the EpiTyper

was in agreement with observations from the array with hypomethylation observed in

HF relative to LF groups (Figure 3.5A). To determine whether hypomethylation at this

fDMR was indeed associated with the functional activity of the distal ZFP57 gene, we

measured both mRNA gene expression and histone acetylation in the same CD4+

samples that were analysed by microarray. A TaqMan PCR amplicon and an in-house

histone H3/H4 acetylation assay targeted to the transcriptional start site of ZFP57 were

designed. A comparison of HF and LF groups revealed increased levels of histone H3

and H4 acetylation at ZFP57, a marker of increased transcriptional activity (Figure

3.5B). In general absolute mRNA expression levels were low but relative differences

were in the order of 4-fold between groups which was statistically significant. (Figure

3.5C). Collectively these lines of evidence suggest a functional role for observed

differences in DNA methylation associated with ZFP57.

Figure 3.5 This figure shows validation data of chromosome 6 fDMR and functional assessment of ZFP57 in CD4+ cells. (A) Consistent with findings from the Illumina array, data from Sequenom EpiTyper platform showing the trend of lower methylation in HF group and higher methylation in LF group. (B) mRNA expression of ZFP57 in CD4+ cells shows an upregulation of the gene in HF group. (C) Levels of H3 and H4 acetylation at ZFP57 promoter region are higher in the HF group indicating a more open chromatin state in this group. Details of the primer sequences are given in the Appendix 2. fDMR; Folate sensitive differentially methylated region, HF; High folate, LF; Low folate.

61

Assessment of reproducibility in an independent set of samples

An independent sample population was next drawn from the source cohort in order to

determine the potential reproducibility of the findings from our initial discovery cohort.

We sampled an additional set of 19 samples from the first and third quartiles of the

maternal serum folate distribution curve from the source population. The characteristics

of these additional samples were similar to the discovery cohort including a linear

relationship between maternal serum folate levels and cord serum folate levels (r = 0.76,

P < 0.001) (Figure 3.1B) but with the notable exception that vitamin D levels were not

different in this group (P = 0.2). Both CD4+ and APC populations were purified from

cord blood as described previously. In agreement with data obtained from the discovery

cohort, levels of DNA methylation were consistently lower at the ZFP57 fDMR in

samples from the HF group (Figure 3.6).

Figure 3.6 Sequenom Epityper analysis of DNA methylation at the chromosome 6 fDMR in an independent set of samples (n=19). Data shows mean DNA methylation level at each CpG site with standard errors. Statistics represent paired two-tailed t-test for average DNA methylation level across all 8 CpG sites (assay average). fDMR; folate sensitive differentially methylated region.

62

3.5 Discussion

The folic acid metabolic pathway plays a critical role in fetal development (23) yet its

effects on regulation of fetal DNA methylation and gene expression are incompletely

understood. In this study we investigated the effects of third trimester maternal folate

status on the DNA methylation profile of neonatal immune cells, with a view to

identifying fDMRs in the genome. We began with a hypothesis free discovery approach

with subsequent replication of candidate loci in an independent set of samples. With this

strategy we have identified regions of the methylome that appear sensitive/responsive to

folic acid status in pregnancy.

Using a genome-scale platform we report several interesting features of the methylation

landscape in neonates stratified by folate exposure in gestation. It is noteworthy that

average DNA methylation levels, methylation levels at imprinted and repeat elements of

the genome appear unperturbed. Despite this, our analysis identified seven folate-

sensitive regions of methylation change in either direction. Folate plays a well-known

role as a carrier of one-carbon units that ultimately resulting in providing methyl groups

to methylate DNA, hence higher folate levels are expected to increase the levels of

DNA methylation. However, many studies, both in humans and animals, have reported

that high folate is associated with hypo- and hyper methylation either globally or in a

gene-specific manner (3, 11, 24). Ontology enrichment analysis of fDMRs suggests

these differences affect important genes involved in the regulation of DNA methylation

during fetal development. Principal among these is ZFP57, a gene that plays a central

role in regulation and maintenance of imprinting-associated DNA methylation (25, 26).

Both data from the microarray and the EpiTyper platform suggest HF exposure was

associated with hypomethylation of a 923bp region localized within a CpG dense

cluster 3kb upstream of the currently annotated ZFP57 transcription start site. Analysis

of histone acetylation profiles in the promoter of ZFP57 and gene expression levels

suggest hypomethylation at this fDMR is associated with markers of increased

transcriptional activity. It is therefore tempting to speculate that a compensatory

mechanism or a negative feed-back loop may exist to regulate the activity of the ZFP57

gene in response to folate exposure to stabilize DNA methylation levels at imprinting

control regions. This assertion is based on the observation that imprinted regions

targeted by the Illumina array did not differ according to folate status. However, within

what range of folate exposure that this mechanism is biologically plausible and what

other factors that may interfere with this compensatory mechanism is yet to be explored.

63

Accordingly, a recent study revealed that ZFP57 is expressed in low but detectable

levels in adult tissues including peripheral blood mononuclear cells and suggests that

ZFP57 may have other functions beyond imprinting establishment and maintenance

(27). More recently, a study focusing on genes down-regulated in association with

embryonic stem cell differentiation in mice, identified ZFP57-mediated IGF2 up

regulation in anchorage-independent growth of cancer cells (28). The authors conclude

that ZFP57 acts as an oncogene in many cancer types. Further studies are needed to

reconcile these findings.

Our two-cell type screen suggests many of the fDMRs identified in this study are

present more broadly throughout the hematopoietic compartment, and given that APCs

and CD4+ represent distinct lineages, suggest that folate driven effects on the epigenome

have originated at the common hematopoietic progenitor stage. In addition we identified

a small number of cell-specific CpG sites associated with folic acid exposure, however

it is likely we are underpowered to characterize these smaller effects in sufficient detail.

Analysis of a second sample set drawn from the source population suggests the findings

reported here are likely to be reproducible to future investigators. Despite this, we have

used an extremes of exposure design in which candidates were selected on the basis of

serum folate levels, and therefore we cannot extend the effect sizes reported here to the

general population. Nevertheless this study reports novel targets that represent viable

research avenues for more large-scale population based studies in the future. These will

be important to determine the extent to which periconceptional folate exposure modify

the disease risk in offspring.

64

3.6 References

1. Foley DL, Craig JM, Morley R, Olsson CA, Dwyer T, Smith K, et al. Prospects

for epigenetic epidemiology. Am J Epidemiol. 2009 Feb 15;169(4):389-400.

2. Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, et al. In

utero supplementation with methyl donors enhances allergic airway disease in

mice. J Clin Invest. 2008 Oct;118(10):3462-9.

3. Cho CE, Sanchez-Hernandez D, Reza-Lopez SA, Huot PS, Kim YI, Anderson

GH. High folate gestational and post-weaning diets alter hypothalamic feeding

pathways by DNA methylation in Wistar rat offspring. Epigenetics. 2013

Jul;8(7):710-9.

4. Waterland RA, Jirtle RL. Early nutrition, epigenetic changes at transposons and

imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition.

2004 Jan;20(1):63-8.

5. Lumley J, Watson L, Watson M, Bower C. Periconceptional supplementation with

folate and/or multivitamins for preventing neural tube defects. Cochrane Database

Syst Rev. 2001(3):CD001056.

6. Kiefte-de Jong JC, Timmermans S, Jaddoe VW, Hofman A, Tiemeier H, Steegers

EA, et al. High circulating folate and vitamin B-12 concentrations in women

during pregnancy are associated with increased prevalence of atopic dermatitis in

their offspring. J Nutr. 2012 Apr;142(4):731-8.

7. Dunstan JA, West C, McCarthy S, Metcalfe J, Meldrum S, Oddy WH, et al. The

relationship between maternal folate status in pregnancy, cord blood folate levels,

and allergic outcomes in early childhood. Allergy. 2012 Jan;67(1):50-7.

8. Haberg SE, London SJ, Stigum H, Nafstad P, Nystad W. Folic acid supplements

in pregnancy and early childhood respiratory health. Arch Dis Child. 2009

Mar;94(3):180-4.

9. Whitrow MJ, Moore VM, Rumbold AR, Davies MJ. Effect of supplemental folic

acid in pregnancy on childhood asthma: a prospective birth cohort study. Am J

Epidemiol. 2009 Dec 15;170(12):1486-93.

10. Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, Lindemans J, Siebel C,

Steegers EA, et al. Periconceptional maternal folic acid use of 400 microg per day

is related to increased methylation of the IGF2 gene in the very young child. PLoS

One. 2009;4(11):e7845.

11. Hoyo C, Murtha AP, Schildkraut JM, Jirtle RL, Demark-Wahnefried W, Forman

MR, et al. Methylation variation at IGF2 differentially methylated regions and

65

maternal folic acid use before and during pregnancy. Epigenetics. 2011

Jul;6(7):928-36.

12. Girardot M, Feil R, Lleres D. Epigenetic deregulation of genomic imprinting in

humans: causal mechanisms and clinical implications. Epigenomics. 2013

Dec;5(6):715-28.

13. Chang H, Zhang T, Zhang Z, Bao R, Fu C, Wang Z, et al. Tissue-specific

distribution of aberrant DNA methylation associated with maternal low-folate

status in human neural tube defects. J Nutr Biochem. 2011 Dec;22(12):1172-7.

14. Fryer AA, Emes RD, Ismail KM, Haworth KE, Mein C, Carroll WD, et al.

Quantitative, high-resolution epigenetic profiling of CpG loci identifies

associations with cord blood plasma homocysteine and birth weight in humans.

Epigenetics. 2011 Jan;6(1):86-94.

15. Binder AM, Michels KB. The causal effect of red blood cell folate on genome-

wide methylation in cord blood: a Mendelian randomization approach. BMC

Bioinformatics. 2013;14:353.

16. Barker DJ. In utero programming of chronic disease. Clin Sci (Lond). 1998

Aug;95(2):115-28.

17. Zaghouani H, Hoeman CM, Adkins B. Neonatal immunity: faulty T-helpers and

the shortcomings of dendritic cells. Trends Immunol. 2009 Dec;30(12):585-91.

18. Reinius LE, Acevedo N, Joerink M, Pershagen G, Dahlen SE, Greco D, et al.

Differential DNA methylation in purified human blood cells: implications for cell

lineage and studies on disease susceptibility. PLoS One. 2012;7(7):e41361.

19. Martino DJ, Tulic MK, Gordon L, Hodder M, Richman T, Metcalfe J, et al.

Evidence for age-related and individual-specific changes in DNA methylation

profile of mononuclear cells during early immune development in humans.

Epigenetics. 2011 Sep 1;6(9).

20. Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, et

al. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina

Infinium HumanMethylation450 microarray. Epigenetics. 2013 Feb;8(2):203-9.

21. Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by

surrogate variable analysis. PLoS Genet. 2007 Sep;3(9):1724-35.

22. Jaffe AE, Murakami P, Lee H, Leek JT, Fallin MD, Feinberg AP, et al. Bump

hunting to identify differentially methylated regions in epigenetic epidemiology

studies. Int J Epidemiol. 2012 Feb;41(1):200-9.

66

23. Gueant JL, Namour F, Gueant-Rodriguez RM, Daval JL. Folate and fetal

programming: a play in epigenomics? Trends Endocrinol Metab. 2013

Jun;24(6):279-89.

24. Sie KK, Li J, Ly A, Sohn KJ, Croxford R, Kim YI. Effect of maternal and

postweaning folic acid supplementation on global and gene-specific DNA

methylation in the liver of the rat offspring. Mol Nutr Food Res. 2013

Apr;57(4):677-85.

25. Zuo X, Sheng J, Lau HT, McDonald CM, Andrade M, Cullen DE, et al. Zinc

finger protein ZFP57 requires its co-factor to recruit DNA methyltransferases and

maintains DNA methylation imprint in embryonic stem cells via its transcriptional

repression domain. J Biol Chem. 2012 Jan 13;287(3):2107-18.

26. Quenneville S, Verde G, Corsinotti A, Kapopoulou A, Jakobsson J, Offner S, et

al. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide

to affect chromatin and DNA methylation of imprinting control regions. Mol Cell.

2011 Nov 4;44(3):361-72.

27. Plant K, Fairfax BP, Makino S, Vandiedonck C, Radhakrishnan J, Knight JC. Fine

mapping genetic determinants of the highly variably expressed MHC gene ZFP57.

Eur J Hum Genet. 2013 Nov 6.

28. Tada Y, Yamaguchi Y, Kinjo T, Song X, Akagi T, Takamura H, et al. The stem

cell transcription factor ZFP57 induces IGF2 expression to promote anchorage-

independent growth in cancer cells. Oncogene. 2014 Jan 27.

67

Chapter 4

Paper 2

Folate status as a modifier of H3/H4 histone acetylation marks at

allergy-associated genes in human neonatal CD4+ T-cells

Hani Harb1*, Manori Amarasekera2*, Sarah Ashley3, Meri K. Tulic2,4, Petra Ina

Pfefferle1, Daniel P. Potaczek1 , Harald Renz1 , David Martino3,5, Dörthe A. Kesper1* &

Susan L. Prescott2*

1 Institute of Laboratory Medicine, Pathobiochemistry and Molecular Diagnostics,

Philipps University Marburg, Germany 2 School of Paediatrics and Child Health, University of Western Australia, Perth,

Australia 3 Murdoch Children’s Research Institute, Melbourne, Australia. 4 Tolerance Immunitaire, Université de Nice Sophia-Antipolis, Hôpital de l’Archet,

Nice, France 5 University of Melbourne, Melbourne, Australia

*HH and MA contributed equally to this work and share first authorship. DK and SP

contributed equally to this work and share senior authorship.

All authors are members of inFLAME (International Inflammation Network) of the

Worldwide University Network (WUN).

68

4.1 Abstract

In animal models, exposure to high maternal folate can modify developmental

programing of the fetal immune system and the risk of allergic diseases, through

epigenetic changes. While effects on the neonatal methylome have been investigated

(Chapter 3), little is known about effects on histone marks which are also sensitive to in

utero nutritional exposures. The aim of this study was to determine if variations in folate

exposure during pregnancy modulate histone acetylation of genes associated with

allergic inflammation. An in-house method was established and validated which allows

quantitative assessment of histone acetylation in candidate genes in CD4+ T-cells.

Using this protocol, histone 3 (H3) and histone 4 (H4) acetylation levels were analysed

in genes associated with allergy development in neonates exposed to high folate (HF,

n=11) and low folate (LF, n=12) in pregnancy. Baseline mRNA expression levels and

allergen and mitogen stimulated cytokine responses were measured in CD4+ T-cells and

cord blood mononuclear cells (CBMC) respectively. H3 and H4 acetylation at GATA3

and H4 acetylation at IL9 were significantly higher in neonates born to mothers with

high serum folate levels. However, there were no significant differences in baseline

mRNA expression of the corresponding genes between the folate groups indicating that

they might represent loci poised for increased transcriptional activity in response to

activation-induced signals. CBMC cytokine profiles did not reveal significant

differences between the groups, however, generally large sample sizes are required to

detect functional differences between groups. In conclusion, maternal serum folate

levels are likely influencing the developing fetal immune system by modifying histone

acetylation marks. High folate levels are associated with transcriptional permissive

chromatin state at allergy-associated loci and might thus affect disease development in

later life. The established protocol can now be employed in other cohort studies to

measure potential differences on the level of acetylation in genes of interest.

69

4.2 Introduction

In utero epigenetic variation has emerged as a key mechanism underpinning different

patterns of fetal ‘programming’ in response to variable environmental cues (1). While

this appears intended to facilitate adaption to the environmental context for short-term

survival, environment-induced epigenetic variation may also adversely influence the

susceptibility to disease later in life (2,3). Understanding environmental modulation of

the epigenetic programing during early development may lead to a better understanding

of both the developmental origins and the prevention of disease. Early dietary patterns,

and specific nutrients, have been a logical area of focus. In particular, there has been

growing interest in the role of nutrients such as folate, which have plausible epigenetic

effects. Folate is a key mediator in one-carbon metabolism that ultimately provides

methyl groups for DNA methylation (4, 5), however, its role in other aspects of

chromatin modification is less well-known.

Maternal folic acid supplementation is recommended pre-conception and in early

pregnancy to reduce the risk of neural tube defects (6). However, many women continue

to consume higher than necessary doses of folic acid later in pregnancy, well beyond the

window of risk for birth anomalies (7). Studies in mice provided compelling evidence

that supplementation with methyl donors in pregnancy (including folic acid) has

epigenetic effects on asthma-associated genes in the offspring, and increases the allergic

phenotype in the offspring (4). Subsequent observational studies in humans have also

linked folate intake with a higher risk of allergic diseases (7-9) although these findings

have not been entirely consistent (10, 11).

To explore this further, we recently conducted a genome-wide DNA methylation

analysis in immune cells isolated from neonatal cord blood, selected on the basis of in

utero folate exposure (12). Using a conventional ‘extremes of exposure’ design we

identified several regions of the neonatal methylome that appear to be sensitive/

responsive to folate status. Specifically we observed hypomethylation of a 923bp region

3kb upstream of the ZFP57 transcript, a regulator of DNA methylation during

development in neonatal immune cells. However DNA methylation machinery appeared

to be refractory to changes in folate status at allergy-associated genes in this study

population. Dietary availability of methyl donors – in addition to its effects on DNA

methylation – also changes the pattern of histone modifications (13). Therefore, we

hypothesised that folate exposure during pregnancy may have effects on H3/H4

70

acetylation marks and was examined in the same cohort of neonates. Here we developed

and validated a new chromatin immunoprecipitation method, in order to compare

acetylation marks in neonates exposed to high versus low folate levels in pregnancy.

4.3 Methods

Selection of the study population

Details of selection of the study population have been published elsewhere (12). In

brief, 23 neonates were selected from a large birth cohort according to their levels of

folate exposure. The HF (n=11) and LF (n=12) groups were defined according to the

first and third quartiles from the distribution of maternal serum folate levels in

conventional ‘extremes of exposure’ design. All study procedures were carried out in

accordance with full institutional ethics approval (by the Princess Margaret Hospital

Ethics Committee).

Serum folate measurement

Folate levels were measured in maternal and cord serum using a competitive

immunoassay (Immulite 2000; Siemans Medical Solutions Diagnostics, Flanders, NJ,

USA).

Establishment and validation of chromatin immunoprecipitation method

We developed this protocol using CD4+ T-cells from adult volunteers that was approved

by the ethics committee of the Medical Faculty of the Philipps University Marburg,

Germany.

Preparation of CD4+ T-cell chromatin for ChIP

CD4+ T-cells were isolated from CBMC using a two-stage isolation strategy with

magnetic DYNAL beads (Invitrogen). To crosslink histones with DNA, cells were fixed

at RT using 1% formaldehyde for 8 min. The reaction was stopped using 55 µl of 2.5 M

glycine. The cross-linked chromatin was treated with 5M sodium butyrate for 3 min and

then centrifuged at 8000 rpm at room temperature (RT) for 5 min to enhance the

binding of the acetyl group to histones. The chromatin was then incubated with lysis

buffer I at RT for 20 min. After centrifugation at 8 000 rpm at RT for 5 min, the

chromatin was incubated with lysis buffer II containing 1% sodium dodecyl sulfate

(SDS) at RT for 5 min followed by 3 min on ice. The chromatin was then placed

directly in the power-up bioruptor (Diagenode, Belgium) for 30 cycles, where each

71

cycle was 30” on and 30 “off (high power). Afterwards, the chromatin was centrifuged

for 15 min at 28000 rpm to remove any debris that may interfere with the process. The

supernatant was transferred to a new reaction tube and diluted to an SDS concentration

of 0.1%.

Chromatin Immunoprecipitation assay

Sepharose beads (GE Healthcare Bio-sciences AB) were washed once with lysis buffer

II and centrifuged at 1800 rpm for 2 min. After removing the supernatant, the beads

were blocked by incubating overnight at 4oC with 1 mg/ml BSA and 400 µg of salmon

sperm DNA. After the incubation, the beads were centrifuged at 1800 rpm for 5 min and

washed once with 5 ml of lysis buffer II. 30 µl of the beads per immunoprecipitation

(IP) per sample were transferred to new reaction tubes and stored until used. 20 µl beads

per antibody per sample were then added to the chromatin and incubated at 4oC for 2

hours to remove non-specific chromatin bound to Sepharose beads (pre-clearing 1).

After the incubation, supernatant with pre-cleared chromatin was transferred to new 1.5

ml reaction tubes.

The remaining beads were mixed with a combination of 500 µl of lysis buffer II and 1

µg of non-specific IgG antibody (Abcam) per sample. The beads bound to non-specific

antibody were incubated at 4oC for 1 hour. Subsequently, the beads coupled with IgG

antibody were centrifuged at 1800 rpm for 5 min and washed 3 times each with 5 ml of

lysis buffer II and centrifuged at 1800 rpm for 5 min. Next, 20 µl of IgG-coupled beads

were added to the pre-cleared chromatin and incubated at 4oC for another 2 hours to

remove chromatin bound to non-specific IgG antibodies (pre-clearing 2). At the end of

the incubation time, the pre-cleared chromatin was centrifuged at 8000 rpm at 4oC for 5

min. The supernatant containing chromatin was transferred to a new reaction tube and

10% of it was set aside for the input control.

H3 or H4 pan acetylation antibodies (4 µg) (Millipore) were added to the chromatin

obtained at the end of the cleaning process and incubated at 4oC overnight along with a

negative control prepared using 0.5 µg of IgG (Abcam). After the overnight incubation,

30 µl of the IgG-coupled beads were added to each IP and incubated at 4oC for 2 hours.

At the end of the incubation period, the beads were centrifuged at 8000 rpm for 2 min

and washed two times with wash buffer I, two times with wash buffer II, three times

with wash buffer III, and finally two times with 1X TE buffer. After discarding the

72

supernatant, 500 µl of the elution buffer were added to the mixture, vortexed and

incubated at RT for 30 min. The mixture was centrifuged at 8000 rpm for 2 min. The

supernatant containing the precipitated chromatin was transferred to a new reaction

tube.

To reverse the crosslinking, the following mixture was added to each IP product as well

as to the input controls: 20 µl of 5 M NaCl, 10 µl of 0.5M EDTA at pH 8, 20 µl of 1M

Tris at pH7.2, 1 µl of Proteinase K (20 mg/mL), and1 µl of RNase A (10 mg/mL). This

mixture ensures detachment of histones from the DNA and denaturation of the rest of

proteins and any remaining RNA. The samples were incubated at 55oC for 3 hours and

then at 65oC overnight. On the following day, DNA was purified using QIAquick kit

according to the manufacturer’s protocol. All incubation steps were performed on an

overhead rotator. Purified DNA was used in real time polymerase reaction to measure

the enrichment of different promoter regions of genes of interest. All primers are listed

in Appendix 2. The enrichment was calculated using the following formula: %

Enrichment = 100*2[(CTinput – 3.3) - CTsample] .The per cent enrichment of the negative

control (IgG) was subtracted from this value and was normalized to the house keeping

gene (HKG), RPL32 as the ratio of % Enrichment of the gene of interest to %

Enrichment to the HKG. We used RPL32 as the house keeping gene which is always

transcriptionally active and should always be associated with acetylated histone H3 and

H4. MS4A2 encodes a gene that is transcriptionally silent in CD4+ T cells and was used

as a negative control.

Quantitative PCR

First-strand cDNA was generated by reverse transcription of 500 ng of total RNA per

sample with random hexamers using SuperScript VILO (Invitrogen) according to the

manufacturer’s instructions. cDNA was diluted 1:5 in RNAse free waster for gene

expression analysis. Relative quantitation of gene expression was performed using pre-

designed TaqMan assays (Applied Biosystems) for target genes on the ABI 7300 real-

time PCR system in triplicate. All samples were normalized to the house-keeping gene.

Assessment of immune responses

Cryopreserved stocks of CBMC were thawed in RPMI media (Invitrogen, CA, USA)

and treated with DNAseI (5µg/ml) for 10 minutes to digest extracellular DNA. Cells

were washed and resuspended to a concentration of 1x106 cells/ml in appropriate culture

73

media and seeded in 96-well polystyrene plates (Thermo Scientific, MA, USA). For

assessment of immune responses, CBMC were cultured alone or with optimised doses

of house dust mite (HDM; 40µg/ml; Greer, NC, USA), ovalbumin (OVA; 100µg/ml;

Sigma Aldrich, Dartford, UK) or mitogen phytohaemaglutinin (PHA; 1µg/ml; H16,

Murex Biotech Ltd, Dartford, UK) for 48 hours in AIM-V media plus β-

mercaptoethanol (4 x 10-5mol/l). Lymphocyte cultures were maintained in 5% CO2 at 37

°C before supernatants were collected and stored at -20 °C for subsequent quantification

of cytokines (IL-5, IL-13, IFN-γ and TNF) with BioPlex200 system (Bio-Rad

Laboratories, NSW, Australia). The lower limit of detection was 3pg/ml for all the

assays.

Statistical analysis

Qualitative variables were compared by chi-square test. Relationship between two

continuous parameters was analysed using Spearman rank correlation coefficient. Initial

comparisons of quantitative variables between the two folate groups were made by

Mann-Whitney U-test. Adjustment analysis was conducted by multiple linear

regression, in which continuous variables having distribution other than normal

according to Shapiro-Wilk test (positively skewed in all cases) were approximated to

normality by square-root transformation before entering the respective model. All

statistical analyses were done using SPSS 21 software (IBM, Armonk, NY, USA).

4.4 Results

Typically Chromatin immunoprecipitation (ChIP) assays have a high requirement for

large number of freshly isolated cells. This has limited the use of this assay primarily to

cell lines rather than primary cell analysis. To address this, we developed and validated

a novel histone acetylation assay that is amenable to analysis of patient samples in a

clinical setting. We targeted this approach to specific genes previously associated with

the development of allergy. We validated this approach in a study involving 10 non

atopic healthy adult volunteers (male: female=1:1). Here we report the key features of

the protocol that influence the optimum output of the ChIP assay for human CD4+ T-

cells.

As non-activated CD4+ T- cells have a compact chromatin structure, and are resistant to

sonication (14), relatively high SDS concentrations and prolonged sonication has been a

requirement for successful fragmentation of the chromatin. Nevertheless, we were able

74

to demonstrate that pre-incubation of the cells with 1% SDS and sonication of the

chromatin in 1% SDS for 30 min (30 sec on 30 sec off at high power) can produce

chromatin with a fragment length of 100-1000bp (Figure 4.1) which is a prerequisite

for the initial steps of the assay.

In a titration experiment we quantified the lower limits of starting material necessary for

this assay. The lowest number of cells that can be used for the method is 1x105 cells per

assay (Figure 4.2A), below this number the ChIP loses its sensitivity. With this starting

number of cells, we identified the lowest enrichment measureable by the used method

for analysed genes in comparison to the positive and negative controls (Appendix 2).

Additionally, samples were frozen and thawed to estimate the freeze-thawing effect on

the enrichment percentage in comparison to freshly isolated and processed samples.

There was a trend towards lower enrichment after freezing the chromatin at some loci,

but this was not significant (Figure 4.2B) indicating that frozen samples can be used for

this assay without affecting the yield.

Figure 4.1 Outline of the ChIP assay and shearing efficiency. (A) Flow Chart outlining the main steps in ChIP method. (B) Shearing efficiency of chromatin at different sodium dodecyl sulfate (SDS) concentrations.

75

There was a significant decrease in the enrichment percentage at RPL32 (positive

control) and IL4 promoter in the 24 h RT chromatin in comparison with fresh chromatin

indicating that the samples should be analysed or frozen immediately (Table 4.1). There

was no difference in the enrichment percentage between freshly isolated and 7 or 30

days frozen chromatin or cells, thus, stored samples can be readily utilised for batch

analysis.

Figure 4.2 Lower limit of quantification and freeze-thawing effect of chromatin. (A) The lowest number of cells that can be used for this method is 1x105 cell per assay. (B) There was no significant effect of freezing on the ChIP assay.

76

Table 4.1 The effect of temperature and long-term storage on per cent enrichment of H3 and H4 histone acetylation ± SEM

Having established the ChIP assay and validating this methodology, we next applied the

technique to our clinical folate cohort to address our primary research question. We

compared H3 and H4 acetylation levels between the HF and LF groups for a panel of

key allergy-associated genes.

The characteristics of the neonates selected for this study have been published

elsewhere (12). In summary, the maternal serum folate measurement at the recruitment

visit were HF= 74.6 nmol/l, LF=16.8 nmol/l (P < .001) and cord serum levels were

HF=78.2nmol/l, LF= 36.4 nmol/l (P<.001) with clear linear relationship between

maternal serum folate and cord blood folate levels in the sample population (R=.75,

P<.001).

The level of H3 and H4 acetylation at the GATA3 locus and H4 at the IL9 locus were

higher in neonate in HF group (Figure 4.3). Additionally, there was a consistent

tendency for increased acetylation of H3 histone at the IL9 locus although this did not

reach full significance. To determine whether increased acetylation level, a marker of

increased transcriptional activity is associated with functional activity of the gene, we

measured mRNA gene expression of the loci that were significantly differently

acetylated between groups. However, there were no significant differences in baseline

mRNA expression of the analysed genes between the folate groups (data not shown). In

order to determine the functional consequences of differences in the acetylation profile

on cytokine production, bulk cord blood mononuclear cells were stimulated with house

dust mite (HDM), ovalbumin (OVA) and phytohaemaglutinin (PHA). We did not

observe any difference in cytokine profile between folate groups (data not shown).

77

Figure 4.3 Levels of H3 (A) and H4 (B) histone acetylation at key genes associated with allergic inflammation in neonatal CD4+ T-cells. All data are presented as mean relative enrichment to the house keeping gene, RPL32 ± SEM. (*P < .05, **P< 0.01)

4.5 Discussion

Recent concerns over the biological effects of in utero exposure to high levels of folate

as a potential risk factor for allergic diseases have been so far limited to animal models

(4) and a small number of human observational studies (7-11). In a recent genome-wide

methylation study, we have provided evidence for exposure to folate has effects on the

regulation of DNA methylation during fetal development (12), however, the exposure

did not appear to modulate the offspring DNA methylation at loci associated with

development of allergic inflammation. Using a candidate approach, we extended the

analysis to examine histone acetylation as an underlying mechanism for possible effects

of gestational folate exposure on the patterns of immune response in the neonate. In

particular, we examined isolated CD4+T-cells, which are central determinants of the

relative polarisation of adaptive effector responses and focused on loci which are known

to be involved in the development of allergic disease. Importantly, by using purified cell

populations in this study, we were able to avoid potential confounding effects of cell

heterogeneity that may affect the measured epigenetic changes (15, 16).

Higher acylation levels at GATA3 and IL9 loci were observed in neonates who had been

exposed to relatively higher levels of folate. GATA-3 directs CD4+ T-cell polarisation

towards Th2 pathway (17) and IL-9, initially described as a Th2 cytokine but now

shown to be produced by an array of Th subsets, is associated with allergic

inflammation and autoimmune diseases (18, 19). The data suggest that exposure to high

gestational folate is associated with more transcriptionally permissive chromatin at

78

allergy-associated genes in neonatal CD4+ T-cells, thus potentially program the

immune profile towards an allergic phenotype. The level of gene expression in resting

cells did not reflect the differences in histone acetylation; however this does not rule out

the possibility that activation-induced expression of these targets may be altered

between groups and should be addressed in future studies. The cytokine responses of

CBMC also did not reveal any difference between the folate groups. However, the

sample size in this study is smaller than generally required to detect significant

functional differences. Collectively these findings suggest that maternal folate status

may influence the fetal immune trajectory by modulating the histone acetylation marks

that may affect the disease development in later life.

This is the first study that investigated variation in histone acetylation in neonatal

immune cells according to in utero folate exposure. The folate-sensitive targets we have

identified in this study provides the future direction upon which more detailed

investigation can be undertaken particularly with large cohorts, for which, the

established protocol can now be employed.

79

4.6 References

1. Gluckman PD, Hanson MA. Developmental origins of disease paradigm: a

mechanistic and evolutionary perspective. Pediatr Res. 2004;56(3):311-7.

2. Barker DJ. In utero programming of chronic disease. Clin Sci (Lond).

1998;95(2):115-28.

3. Barker DJ. The intrauterine environment and adult cardiovascular disease. Ciba

Found Symp. 1991;156:3-10; discussion -6.

4. Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, et al. In

utero supplementation with methyl donors enhances allergic airway disease in

mice. J Clin Invest. 2008;118(10):3462-9.

5. Cho CE, Sanchez-Hernandez D, Reza-Lopez SA, Huot PS, Kim YI, Anderson

GH. High folate gestational and post-weaning diets alter hypothalamic feeding

pathways by DNA methylation in Wistar rat offspring. Epigenetics.

2013;8(7):710-9.

6. Jagerstad M. Folic acid fortification prevents neural tube defects and may also

reduce cancer risks. Acta Paediatr. 2012;101(10):1007-12.

7. Dunstan JA, West C, McCarthy S, Metcalfe J, Meldrum S, Oddy WH, et al. The

relationship between maternal folate status in pregnancy, cord blood folate levels,

and allergic outcomes in early childhood. Allergy. 2012;67(1):50-7.

8. Kiefte-de Jong JC, Timmermans S, Jaddoe VW, Hofman A, Tiemeier H, Steegers

EA, et al. High circulating folate and vitamin B-12 concentrations in women

during pregnancy are associated with increased prevalence of atopic dermatitis in

their offspring. J Nutr. 2012;142(4):731-8.

9. Whitrow MJ, Moore VM, Rumbold AR, Davies MJ. Effect of supplemental folic

acid in pregnancy on childhood asthma: a prospective birth cohort study. Am J

Epidemiol. 2009;170(12):1486-93.

10. Bekkers MB, Elstgeest LE, Scholtens S, Haveman-Nies A, de Jongste JC,

Kerkhof M, et al. Maternal use of folic acid supplements during pregnancy, and

childhood respiratory health and atopy. Eur Respir J. 2012;39(6):1468-74.

11. Magdelijns FJ, Mommers M, Penders J, Smits L, Thijs C. Folic acid use in

pregnancy and the development of atopy, asthma, and lung function in childhood.

Pediatrics. 2011;128(1):e135-44.

12. Amarasekera M, Martino D, Ashley S, Harb H, Kesper D, Strickland D, et al.

Genome-wide DNA methylation profiling identifies a folate-sensitive region of

80

differential methylation upstream of ZFP57-imprinting regulator in humans.

FASEB J. 2014.

13. Pogribny IP, Tryndyak VP, Muskhelishvili L, Rusyn I, Ross SA. Methyl

deficiency, alterations in global histone modifications, and carcinogenesis. J Nutr.

2007;137(1 Suppl):216S-22S.

14. Rawlings JS, Gatzka M, Thomas PG, Ihle JN. Chromatin condensation via the

condensin II complex is required for peripheral T-cell quiescence. EMBO J.

2011;30(2):263-76.

15. Adalsteinsson BT, Gudnason H, Aspelund T, Harris TB, Launer LJ, Eiriksdottir

G, et al. Heterogeneity in white blood cells has potential to confound DNA

methylation measurements. PLoS One. 2012;7(10):e46705.

16. Reinius LE, Acevedo N, Joerink M, Pershagen G, Dahlen SE, Greco D, et al.

Differential DNA methylation in purified human blood cells: implications for cell

lineage and studies on disease susceptibility. PLoS One. 2012;7(7):e41361.

17. Wilson CB, Westall J, Johnston L, Lewis DB, Dower SK, Alpert AR. Decreased

production of interferon-gamma by human neonatal cells. Intrinsic and regulatory

deficiencies. J Clin Invest. 1986;77(3):860-7.

18. Pan HF, Leng RX, Li XP, Zheng SG, Ye DQ. Targeting T-helper 9 cells and

interleukin-9 in autoimmune diseases. Cytokine & growth factor reviews.

2013;24(6):515-22.

19. Brough HA, Cousins DJ, Munteanu A, Wong YF, Sudra A, Makinson K, et al. IL-

9 is a key component of memory TH cell peanut-specific responses from children

with peanut allergy. J Allergy Clin Immunol. 2014;134(6):1329-38 e10.

81

Chapter 5

Paper 3

Epigenome-wide analysis of neonatal CD4+ T-cell DNA methylation

sites potentially affected by maternal fish oil supplementation

Manori Amarasekera1, Paul Noakes1, Deborah Strickland2, Richard Saffery3,4, David J

Martino3,4,5*, Susan L Prescott1,2*

1School of Paediatrics and Child Health, University of Western Australia, Perth,

Australia 2Telethon Kids Institute, Perth, Australia 3Murdoch Children’s Research Institute, Melbourne, Australia 4Department of Paediatrics, University of Melbourne, Melbourne, Australia

5Centre for Food and Allergy Research, Melbourne, Australia

*DM and SLP contributed equally to this work and share senior authorship.

All authors are members of inFLAME (International Inflammation Network) of the

Worldwide University Network.

82

5.1 Abstract

Supplementation of fish oil rich in omega-3 polyunsaturated fatty acids (n-3 PUFA)

during pregnancy has been shown to confer favourable health outcomes in the offspring.

In a randomised controlled trial, we have previously shown that n-3 PUFA

supplementation in pregnancy was associated with modified immune responses and

some markers of immune maturation. However, the molecular mechanisms underlying

these heritable effects are unclear. To determine whether the biological effects of

maternal n-3 PUFA supplementation are mediated through DNA methylation we

analysed the CD4+ T-cells purified from cryo-banked cord blood samples from

previously conducted clinical trial. Of the 80 mother-infant pairs completed the initial

trial, cord blood samples of 70 neonates were available for genome-wide DNA

methylation profiling. Comparison of purified total CD4+ T-cell DNA methylation

profiles between the supplement and control groups did not reveal any statistically

significant differences in CpG methylation, at the single-CpG or regional level. Effect

sizes among top-ranked probes were less than 5% and did not warrant further

validation. Tests for association between methylation levels and key n-3 PUFA

parameters docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) or total n-3

PUFAs were suggestive of dose-dependent effects, but these did not reach genome-wide

significance. Our analysis of the microarray data did not suggest strong modifying

effects of in utero n-3 PUFA exposure on CD4+ T-cell methylation profiles, and no

probes on the array met our criteria for further validation. Other epigenetic mechanisms

may be more relevant mediators of functional effects induced by n-3 PUFA in early life.

83

5.2 Introduction

Supplementation of polyunsaturated fatty acids (PUFAs) during pregnancy and during

early development has been shown to be potentially beneficial in preventing allergic (1-

3) cardiovascular (4, 5) and metabolic disorders (6) in the offspring. We have

previously shown that supplementation with n-3 PUFA DHA (22:6 n-3) and EPA (20:5

n-3) from 20 weeks of gestation until delivery modified inflammatory immune

responses in neonatal immune cells (1, 7-9). While this dietary intervention affects

many biochemical and physiological properties of cells and organs, the exact molecular

mechanism by which n-3 PUFAs exert their multi-system benefits is yet to be fully

elucidated. Specifically, in the context of heritable effects of maternal exposure, the

proposed pathways do not provide a clear molecular link.

There is mounting evidence that dietary factors can change cellular epigenetic marks in

association with more favourable clinical outcomes (10). Dietary PUFAs have also been

shown to regulate gene expression through epigenetic mechanisms (11-13). Treatment

of U937 leukemic macrophage cells with EPA has been shown to increase mRNA

expression of CCAAT/enhancer-binding protein delta with concomitant demethylation

of specific CpG loci in the gene promoter region (11) whereas treatment of M17

neuroblastoma cells with DHA tended to induce histone modifications that are

consistent with active transcription (12). Extending these findings to human cohorts, a

recent study reported 27 differentially methylated CpG sites at biologically relevant

regions that were associated with n-3 PUFA intake using an “extreme phenotypes”

model in a cohort of 185 adults (14). Another study that investigated the association of

n-6 PUFA intake and central obesity in 40 young women found that the level of TNF

promoter methylation of peripheral white cells was associated with n-6 PUFA intake

(15). Of greatest relevance here, emerging data from animal studies support a role for

maternal intake of PUFA in pregnancy in the modulation of offspring epigenetic profile

(16, 17). In a recent study involving pregnant women who had been supplemented with

DHA or placebo from the second trimester until delivery, Lee et al measured the DNA

methylation changes in genes associated with Th1, Th2, Th17, and regulatory T cell

development, as well as LINE1 repetitive elements of cord blood mononuclear cells

(18). Although there were no significant differences in DNA methylation pattern in

genes when comparing the treatment and the placebo groups, overall n-3

supplementation modified effects on methylation in the subgroup of neonates whose

mothers smoked during pregnancy. Specifically in this risk group, n-3 supplementation

84

was associated with differences in methylation levels of LINE1 repetitive element.

Collectively, these data suggests maternal n-3 PUFA intake during pregnancy may

modify the fetal epigenome, and that this may depend on other environmental

exposures.

To determine whether exposure to n-3 PUFA intake during pregnancy alters the fetal

epigenome, we compared whole genome DNA methylation profiles from neonatal

CD4+ T-cells derived from previously carried out randomized controlled trial of

maternal supplementation of fish oil during pregnancy. CD4+ T-cells were chosen as

they play a key role in immune responses and have been shown to be susceptible to in

utero perturbation from environmental exposures (19, 20). We hypothesised that

maternal n-3 PUFA supplementation may modulate epigenetic programming of CD4+

T-cells, and that this may modulate later health outcomes.

5.3 Materials and Methods

Study population

We used samples of the well-established study cohort which was a double-blind

randomised trial of pregnant mothers who were atopic and had clinical allergy (either

allergic rhinitis or asthma) but were otherwise healthy (1). They were randomised to

receive either 3.7g of fish oil (with 56.0% as DHA and 27.7% as EPA) or placebo in

capsules daily from 20 weeks of gestation until delivery. 83 women and their healthy

full-term babies completed the trial. All study procedures were carried out in

accordance with full institutional ethics. Full details of this cohort have been published

elsewhere (1).

Data and blood sample collections

Maternal fasting blood samples were collected at 20 weeks of gestation to determine the

maternal baseline fatty acids levels and at 37 weeks to calculate the increment in fatty

acids levels during the study period. Women also completed a validated semi-

quantitative food frequency questionnaire.

Cord blood samples were collected at the time of birth. Cord blood mononuclear cells

(CBMC) were harvested from blood samples within 12 hours of collection according to

standard protocols (21). Of the completed participants, a total of 70 cord bloods were

available for the present study. Maternal antenatal and socio-demographic factors and

infant clinical outcomes at 1year were also available.

85

Erythrocyte fatty acid composition analysis

Both maternal and cord blood samples were collected and processed according to

standard protocols to determine the fatty acid composition in red cells (22). The fatty

acids were expressed as a percentage of the weight of the total fatty acids measured. The

total sum of n-3 PUFAs (20:5n-3, 22:5n-3, and 22:6n-3) and n-6 PUFAs (18:2n-6,

20:3n-6, 20:4n-6, 22:3n-6, and 22:4n-6) as well as the ratio of n3 to n6 fatty acids were

also expressed.

Isolation of CD4+ T-cells

CD4+ T cells were isolated from CBMC using a two-stage positive isolation strategy

with magnetic DYNAL beads (Invitrogen). CBMC were incubated with CD8+ magnetic

beads as per the recommended protocol (Invitrogen), and the CD8- fraction was then

incubated with CD4+ magnetic beads as per the recommended protocol. Routine purity

tests were conducted by flow cytometry using antibodies CD19-FITC, CD3-PE, CD8-

PerCP, CD11c-PE Cy7, CD4-APC and CD14-APC Cy7 (BD Biosciences) and

appropriate concentration matched istoype controls. CD4+ cell purities ranged from 91-

96% pure. CD4+ T cells were lysed with RLT buffer containing 2-mercaptoethanol

(Qiagen Allprep kit) and stored at -80 ºC until genomic DNA was extracted for

methylation analysis.

Nucleic acid extraction

Genomic DNA was purified from CD4+ T- cells using Qiagen Allprep kits according to

manufacturer’s instructions.

DNA methylation analysis

DNA samples were submitted to the NXT-Dx service facility (Belgium) for bisulphite

conversion and DNA methylation profiling using the Illumina Human Methylation 450k

platform. Samples were randomized across arrays according to treatment group, age and

maternal allergy status. Raw data (iDAT files) were processed using the Minfi package

(23) from the bioconductor project (http://www.bioconductor.org) in the R statistical

environment (http://cran.r-project.org/). The minfi package was used evaluate control

probes on the array and both sample-dependent and assay-dependent control probes

indicated high quality data. Arrays were pre-processed using the stratified quantile

normalization method. Technical bias attributable to different probe chemistries

between Type 1 and Type II probes were adjusted in this procedure. Probes with a

86

detection P-value call >0.01 in 1 or more samples were removed. Probes on the X and

Y-chromosomes were removed to eliminate gender bias. Probes previously

demonstrated to potentially cross-hybridize non-specifically in the genome were

removed (24) Probes containing a polymorphic SNP at the single-base extension site

with a minor allele frequency of <0.05 were removed. The log2 ratio for methylated

probe intensity to unmethylated probe intensity, the M value, was subsequently derived

and used for statistical inference. Beta values were derived from intensities as defined

by the ratio of methylated to unmethylated probes given by B=M/(U/M*100) and were

used to compliment M-value as a measure of effect size. Cluster analysis was used in

conjunction with the chip-wide medians of the methylated and unmethylated channels

to identify any outlying samples, all samples passed QC. The final data set included 424

803 probes.

Data analysis

Following pre-processing a surrogate variable analysis was conducted on normalized

M-values to estimate any residual variation not associated with the primary outcomes of

interest using the method of Storey and Leek (25). This method estimates unwanted

variation empirically from the observed data and latent variables constructed from this

analysis were included in the regression model as covariates to negate their effects. To

identify CpG sites associated with treatment group or total n-3 fatty acid levels, all

probes were regressed on these independent variables and the empirical Bayes method

of Smyth et al (26) was used to compare treatment groups. For continuous variables, we

used the CpGassoc statistical package (27) and included surrogate variables as the

covariate matrix. QQ-plots of P-value distributions and scatterplots were used to

visualise data. Adjustment for multiple testing was performed using the Benjamini

Hochberg method (28) with adjusted P-Values < 0.1 considered significant. Our criteria

for identifying significant hits for further validation studies included a P-value < 0.05

after correction for multiple testing, and an effect size greater than 5%.

5.4 Results

Characteristics of the cohort

Of the total 83 mother-infant pairs who completed the initial randomised controlled

trial, 70 had cord blood available for this epigenome-wide DNA methylation analysis.

Table 5.1 shows the characteristics of the 70 mother-infant pairs; 36 in the fish oil

87

group and 34 in the control group. There were no significant differences in the key

maternal and neonatal parameters between treatment and control groups.

Table 5.1 Population characteristics of the mothers and infants in the cohort

(n=70)

Characteristic

Fish oil

(n=36)

Control

(n=34) P value

Mothers at recruitment Age, (yrs [SE]) 31.1 (0.7) 32.6 (0.6) 0.09

BMI at entry (kg/m2 [SE]) 25.5 (0.8) 25.9 (0.7) 0.726 Asthma (n [%]) 15 (42%) 12 (35%) Allergic rhinitis (n [%]) 21 (58%) 22 (65%) 0.584 Exposed to passive smoke (n [%]) 20 (56%) 19 (56%) 0.811 Infants

Male (n [(%]) 16 (44%) 21 (62%) 0.147 Gestation (days [SE]) 274.5 (1.4) 273.7 (1.4) 0.687 Delivery method

Vaginal (n [(%]) 26 (72%) 27 (79%) Caesarean section (n [(%]) 10 (28%) 7 (21%) 0.483

Neonatal growth parameters Birth weight (g [SE]) 3491 (57.1) 3481 (63.6) 0.906

Length (cm [SE] 50.4 (0.3) 49.7 (0.3) 0.117 Head circumference (cm [SE]) 35.2 (0.2) 34.6 (0.2) 0.065 Fatty acid composition in maternal

erythrocytes at 37 weeks of gestation

eicosapentaenoic acid, EPA (SE) 2.51 (0.11) 0.68 (0.02) < 0.001*

docosahexaenoic acid, DHA (SE) 10.75 (0.33) 6.39 (0.15) < 0.001* Arachidonic acid, AA (SE) 10.37 (0.28) 13.83 (0.16) < 0.001* Total n-3 PUFA (SE)b 22.31 (0.58) 15.98 (0.29) < 0.001* Total n-6 PUFA (SE)c 21.38 (0.53) 28.44 (0.27) < 0.001* Total n-3 to n-6 (SE)d 1.06 (0.03) 0.57 (0.01) < 0.001*

Statistical comparisons by Chi square and Man-Whitney U-test . a Values for fatty acids are expressed as a percentage of total fatty acids. b Sum 20:5, 22:5, 22:3. c Sum 18:2, 20:3, 20:4, 22:3, 22:4. d Ratio [Sum 20:5, 22:5, 22:3] to [18:2, 20:3, 20:4, 22:3, 22:4].

* indicates significant P-values (P < 0.05)

Maternal supplementation with n-3 PUFA modifies fetal red cell parameters

To determine effectiveness of the maternal intervention fatty acid parameters in cord red

blood cells were compared between treatment and intervention group. There was clear

evidence of significantly higher total n-3 (fish oil=17.8; control 13.6; P < 0.001) and

lower total n-6 (fish oil=25.2; control 29.6; P < 0.001) fatty acid levels in treatment

88

group. Key n-3 PUFAs EPA and DHA were also higher in the treatment group while

arachidonic acid a key n-6 PUFA were lower compared to that of the control group

(Figure 5.1). These readouts provided robust evidence that the maternal

supplementation modified fetal lipid profiles.

Figure 5.1 Cord red blood cell fatty acid measurements in fish oil and control groups. Boxplots represent median and range. Statistical analysis by Man-Whitney U-test. EPA-eicosapentaenoic acid, DHA-docosahexaenoic acid, AA- arachidonic acid, total n-3 PUFAs (Sum 20:5, 22:5, 22:3), total n-6 PUFAs (Sum 18:2, 20:3, 20:4, 22:3, 22:4)

Identification of CpG sites that co-associate with n-3 red cell fatty acid levels

To identify CpG sites associated with n-3 supplementation in the neonatal CD4+ T-cell

epigenome, we regressed all probes on neonatal total n-3 red cell fatty acid levels as a

continuous variable in the presence of latent covariates (methods). We did not identify

any CpG sites that were significantly associated with total n-3 levels that survived

multiple testing (Figure 5.2A). We therefore ranked all CpG sites on the array by un-

adjusted P-values and examined the top-ranked CpG sites for evidence of an effect

using scatterplots. The top 10 ranked CpG were examined by plotting methylation

versus total n-3 fatty acid level. A dose-response relationship was apparent among the

CpG sites ranked at the top of the list and these relationships were bi-directional at the

individual CpG level, with some CpG positively associated with n-3 fatty acid levels

89

and others negatively associated with increasing n-3 fatty acid levels (Figure 5.2B, 4

CpG shown for convenience). Individually these CpG sites exhibited a significant

association with total n-3 fatty acid levels although the effect sizes were small (less than

5% change in methylation) (Figure 5.2C) and not significant at the genome scale, and

therefore of unknown biological relevance. We found this to be the case regardless of

whether the data was modelled on cord red cell DHA levels or EPA levels, or whether

covariates were included in the model or not. We filtered the data set to include only the

top 25% most variable probes on the array and repeated the analysis with the same

result (data not shown).

Figure 5.2 Regression analysis of CpG sites that co-associate with cord red cell total n-3 fatty acid levels. (A) qq plot of p-values from the association test between CpGs and total n-3 fatty acids did not deviate from the null hypothesis. (B) Scatterplots of the top four CpGs ranked by p-value for the association with total n-3 fatty acids suggested a dose-response relationship. (C) Histogram of the coefficients from the regression model indicated a small effect size.

Comparison of methylation profiles between treatment and control groups

We next compared genome-wide methylation profiles between treatment and control

groups using multidimensional scaling analysis. Neither genome-wide methylation

profiles, nor the top 5000 most variable probes discriminated the two groups (Figure

5.3A). We compared treatment and control groups for differential methylation in a case

control comparison by regressing all probes on treatment group in the presence of

90

covariates (described in methods). Genome-wide profiles of methylation were not

significantly different (Figure 5.3B), nor did we detect significant differences when we

restricted the analysis to promoter regions or CpG island-associated regions only. We

ranked CpG sites by un-adjusted P-value and examined the top ranked candidates.

There was no clear evidence for a substantial effect of fish oil on the neonatal CD4+ T-

cell methylome (Figure 5.3C). This analysis was repeated for the top 25% most

variable probes, which did not show a significant difference between groups (not

shown). We also report no evidence against the null hypothesis using a sliding windows

analysis to test for regional differences in adjacent CpG sites (not shown).

Figure 5.3 Comparative analysis of DNA methylation profiles between treatment and intervention groups. (A) MDS scaling analysis did not discriminate treatment groups. Samples are indicated by open circles and projected along axes of the first two principal components of the top 5000 most variable CpG sites. (B) qq plot of p-values from the moderated-t test between intervention and control groups did not deviate from the null hypothesis. (C) Boxplots of data from six top-ranked CpG sites for the between-group comparison did not indicate substantive differences in methylation levels. Boxplots show median and range.

5.5 Discussion

This study builds upon an emerging literature suggesting dietary components can

potentially modify epigenetic regulation in humans (10). Foundational work by others

91

(11-14) suggests dietary n-3 PUFAs may alter promoter methylation in blood cells (14).

Here we examined this in the context of supplementation of n-3 PUFA during

pregnancy. In line with the emerging evidence of maternal dietary factors can influence

the neonatal epigenetic profile in modulating immune development (29, 30), we

investigated a potential role of DNA methylation as an underlying mechanism

mediating biological effects of maternal n-3 fish oil supplementation. Our primary

hypothesis was that high maternal supplementation of n-3 PUFA modifies neonatal

CD4+ T-cell DNA methylation profiles. Despite the clear evidence of a modifying effect

of maternal supplementation on red cell lipid profiles, we did not observe substantial

changes in the methylome, at least not at baseline in CD4+ T-cells. Analysis of CpG

sites that co-vary with total n-3 red cell fatty acid levels suggests there may be small

effects at specific CpG sites, but these did not appear to differ in a biologically

meaningful way between the treatment and control groups in this study. Our data do not

support the idea that n-3 PUFA supplementation in pregnancy alters the developing

fetal methylome. Whilst our analysis was restricted to a single purified cell type, we

expect than any effects due to n-3 PUFA exposure during in utero development would

be likely to propagate throughout the hematopoietic system and be present in other

blood cell types as we have reported before (19). Despite this it remains possible that

other blood cell components such as erythrocyte populations may exhibit cell-type

specific effect not observed in CD4+ T-cells. Moreover, investigating the immune

response capacity of CD4+ T-cells was beyond the scope of the present study, and

therefore it remains possible that neonatal fish oil exposure may modify activation-

induced effects on the neonatal DNA methylome. Follow-up studies are required to

address this given our data suggests any effects on the establishment and maintenance

of DNA methylation marks in the neonatal epigenome are likely to be subtle. Other

epigenetic marks such as histone modifications, which in general terms are more

dynamically responsive to environment than DNA methylation, may be key modulators

of n-3 PUFA induced effects on gene transcription. These represent further avenues of

investigation.

To date only a few animal studies have reported the potential effects of maternal PUFA

intake on the fetal epigenome (16, 17). One such study reported differing levels of

FADS2 promoter methylation in the liver tissue from offspring exposed to linoleic acid

during gestation (17). This was replicated in a rat model in which dietary fatty acid

exposure during gestation was associated with differential methylation of 4 CpG

92

dinucleotides within the FADS2 promoter (16). In humans the evidence for these effects

on the neonatal epigenome are lacking, and certainly our own study did not reproduce

these findings in animal models. Similarly, Lee and coworkers analysed a

comparatively larger birth cohort (n=261) and reported that maternal intervention

significantly modulated the LINE1 methylation which is a surrogate marker of global

methylation (18). However, the difference in the methylation level of LINE1 sequences

between supplemented and control group was about 1%, in line with our own findings

in non-repeat regions of the genome and therefore of unknown biological significance.

This is a critical issue in DNA methylation studies as the finite distribution of

methylation measurements is readily amenable to achieving statistical significance, in

situations where the variance distribution across cases and controls is small. However it

is unclear whether these small effects bear any biological relevance at all. DNA

methylation levels mitigate functional effects in specific genomic contexts, and

therefore it is imperative that functionality is established through examination of effect

sizes (with > 10% widely considered to be biologically meaningful), and genomic

context (location of methylation site relative to gene promoter regions, splice sites etc.).

Further research is therefore warranted to uncover the molecular effects of dietary fatty

acid on neonatal immune development. Based on our observations, we anticipate large

cohorts or animal models will be required to address this using genome-wide

association tests and in this respect our study informs future EWAS designs with similar

goals.

Strength of this study is the design in which a priori evidence for a modification effect

on neonatal red blood cell lipid profiles was a driver of our primary investigation.

Moreover the analysis of purified CD4+ T-cells is more homogenous than whole blood

or mononuclear cells, is therefore less subjected to unwanted variation due to cell

composition and is thus expected to provide good sensitivity (31). In summary, we

conducted a hypothesis-free epigenome-wide search to identify genomic regions in

neonatal immune cells that is modulated by maternal n-3 PUFA supplementation. Our

data argues against substantial modifying effects on the neonatal CD4+ T-cell

methylome. This suggests that the well described biological effects of n-3 PUFA are

more likely to be mediated by other epigenetic or post-transcriptional effects that

modulate cellular function.

93

5.6 References

1. Dunstan JA, Mori TA, Barden A, Beilin LJ, Taylor AL, Holt PG, et al. Fish oil

supplementation in pregnancy modifies neonatal allergen-specific immune

responses and clinical outcomes in infants at high risk of atopy: a randomized,

controlled trial. J Allergy Clin Immunol. 2003;112(6):1178-84.

2. Palmer DJ, Sullivan T, Gold MS, Prescott SL, Heddle R, Gibson RA, et al. Effect

of n-3 long chain polyunsaturated fatty acid supplementation in pregnancy on

infants' allergies in first year of life: randomised controlled trial. BMJ.

2012;344:e184.

3. Klemens CM, Berman DR, Mozurkewich EL. The effect of perinatal omega-3

fatty acid supplementation on inflammatory markers and allergic diseases: a

systematic review. BJOG. 2011;118(8):916-25.

4. Skilton MR, Ayer JG, Harmer JA, Webb K, Leeder SR, Marks GB, et al.

Impaired fetal growth and arterial wall thickening: a randomized trial of omega-3

supplementation. Pediatrics. 2012;129(3):e698-703.

5. Forsyth JS, Willatts P, Agostoni C, Bissenden J, Casaer P, Boehm G. Long chain

polyunsaturated fatty acid supplementation in infant formula and blood pressure

in later childhood: follow up of a randomised controlled trial. BMJ.

2003;326(7396):953.

6. Innis SM. Metabolic programming of long-term outcomes due to fatty acid

nutrition in early life. Matern Child Nutr. 2011;7 Suppl 2:112-23.

7. Prescott SL, Barden AE, Mori TA, Dunstan JA. Maternal fish oil supplementation

in pregnancy modifies neonatal leukotriene production by cord-blood-derived

neutrophils. Clin Sci (Lond). 2007;113(10):409-16.

8. Barden AE, Mori TA, Dunstan JA, Taylor AL, Thornton CA, Croft KD, et al.

Fish oil supplementation in pregnancy lowers F2-isoprostanes in neonates at high

risk of atopy. Free Radic Res. 2004;38(3):233-9.

9. Prescott SL, Irvine J, Dunstan JA, Hii C, Ferrante A. Protein kinase Czeta: a novel

protective neonatal T-cell marker that can be upregulated by allergy prevention

strategies. J Allergy Clin Immunol. 2007;120(1):200-6.

10. McKay JA, Mathers JC. Diet induced epigenetic changes and their implications

for health. Acta Physiol (Oxf). 2011;202(2):103-18.

11. Ceccarelli V, Racanicchi S, Martelli MP, Nocentini G, Fettucciari K, Riccardi C,

et al. Eicosapentaenoic acid demethylates a single CpG that mediates expression

94

of tumor suppressor CCAAT/enhancer-binding protein delta in U937 leukemia

cells. J Biol Chem. 2011;286(31):27092-102.

12. Sadli N, Ackland ML, De Mel D, Sinclair AJ, Suphioglu C. Effects of zinc and

DHA on the epigenetic regulation of human neuronal cells. Cell Physiol Biochem.

2012;29(1-2):87-98.

13. Cho Y, Turner ND, Davidson LA, Chapkin RS, Carroll RJ, Lupton JR. Colon

cancer cell apoptosis is induced by combined exposure to the n-3 fatty acid

docosahexaenoic acid and butyrate through promoter methylation. Exp Biol Med

(Maywood). 2014;239(3):302-10.

14. Aslibekyan S, Wiener HW, Havel PJ, Stanhope KL, O'Brien DM, Hopkins SE, et

al. DNA methylation patterns are associated with n-3 fatty acid intake in Yup'ik

people. J Nutr. 2014;144(4):425-30.

15. Hermsdorff HH, Mansego ML, Campion J, Milagro FI, Zulet MA, Martinez JA.

TNF-alpha promoter methylation in peripheral white blood cells: relationship with

circulating TNFalpha, truncal fat and n-6 PUFA intake in young women.

Cytokine. 2013;64(1):265-71.

16. Hoile SP, Irvine NA, Kelsall CJ, Sibbons C, Feunteun A, Collister A, et al.

Maternal fat intake in rats alters 20:4n-6 and 22:6n-3 status and the epigenetic

regulation of Fads2 in offspring liver. J Nutr Biochem. 2013;24(7):1213-20.

17. Niculescu MD, Lupu DS, Craciunescu CN. Perinatal manipulation of alpha-

linolenic acid intake induces epigenetic changes in maternal and offspring livers.

FASEB J. 2013;27(1):350-8.

18. Lee HS, Barraza-Villarreal A, Hernandez-Vargas H, Sly PD, Biessy C,

Ramakrishnan U, et al. Modulation of DNA methylation states and infant immune

system by dietary supplementation with omega-3 PUFA during pregnancy in an

intervention study. Am J Clin Nutr. 2013;98(2):480-7.

19. Amarasekera M, Martino D, Ashley S, Harb H, Kesper D, Strickland D, et al.

Genome-wide DNA methylation profiling identifies a folate-sensitive region of

differential methylation upstream of ZFP57-imprinting regulator in humans.

FASEB J. 2014.

20. Martino D, Joo JE, Sexton-Oates A, Dang T, Allen K, Saffery R, et al.

Epigenome-wide association study reveals longitudinally stable DNA methylation

differences in CD4+ T cells from children with IgE-mediated food allergy.

Epigenetics. 2014;9(7).

95

21. Martino DJ, Tulic MK, Gordon L, Hodder M, Richman T, Metcalfe J, et al.

Evidence for age-related and individual-specific changes in DNA methylation

profile of mononuclear cells during early immune development in humans.

Epigenetics. 2011;6(9).

22. Dunstan JA, Roper J, Mitoulas L, Hartmann PE, Simmer K, Prescott SL. The

effect of supplementation with fish oil during pregnancy on breast milk

immunoglobulin A, soluble CD14, cytokine levels and fatty acid composition.

Clin Exp Allergy. 2004;34(8):1237-42.

23. Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen

KD, et al. Minfi: a flexible and comprehensive Bioconductor package for the

analysis of Infinium DNA methylation microarrays. Bioinformatics.

2014;30(10):1363-9.

24. Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, et

al. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina

Infinium HumanMethylation450 microarray. Epigenetics. 2013;8(2):203-9.

25. Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by

surrogate variable analysis. PLoS Genet. 2007;3(9):1724-35.

26. Smyth GK. Linear models and empirical bayes methods for assessing differential

expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3:Article3.

27. Barfield RT, Kilaru V, Smith AK, Conneely KN. CpGassoc: an R function for

analysis of DNA methylation microarray data. Bioinformatics. 2012;28(9):1280-

1.

28. Benjamini Y HY. Controlling the False Discovery Rate: A Practical and Powerful

Approach to Multiple Testing. JSTOR. 1995;57(1):289-300.

29. Martino DJ, Prescott SL. Silent mysteries: epigenetic paradigms could hold the

key to conquering the epidemic of allergy and immune disease. Allergy.

2010;65(1):7-15.

30. Amarasekera M, Prescott SL, Palmer DJ. Nutrition in early life, immune-

programming and allergies: the role of epigenetics. Asian Pac J Allergy Immunol.

2013;31(3):175-82.

31. Adalsteinsson BT, Gudnason H, Aspelund T, Harris TB, Launer LJ, Eiriksdottir

G, et al. Heterogeneity in white blood cells has potential to confound DNA

methylation measurements. PLoS One. 2012;7(10):e46705.

96

Chapter 6

General Discussion

This thesis describes a unique series of studies that investigate the role of maternal

dietary factors and their capacity to alter the neonatal epigenome. In the broader

context, this thesis aims to establish a mechanistic link between early life exposures, the

developing fetal epigenome and neonatal immune responses, and their role in the risk

for immune diseases. The key findings are summarised and discussed in the context of

existing literature in Section 6.1. Study limitations and suggested directions for future

research are detailed in Section 6.2 and 6.3 respectively and conclusions in Section 6.4.

6.1 Discussion of key findings in the context of existing literature

The general hypothesis underpinning this thesis was that maternal nutritional factors

have the capacity to modify the neonatal epigenome and potentially modify the risk of

immune diseases. We focused on allergic disease as one of the earliest manifestations of

NCD, with a high prevalence in paediatric populations, and thus directed our studies

towards genes and pathways important in the development of allergy. We chose to

study two main dietary factors with well-documented developmental effects that were

readily measurable in pregnant women recruited to this study. The primary aims of this

thesis were to, firstly, examine the variation in the neonatal epigenome according to

levels of in utero folate exposure and, secondly, to investigate epigenetic mechanisms as

the likely mediator of favourable immune outcomes of maternal n-3 PUFA

supplementation during pregnancy.

We took different approaches to test our hypotheses: for the study of DNA methylation

we adopted a hypothesis free approach and utilised cutting edge DNA microarray

technology to discover novel regions of the methylome that may be modified by in

utero exposures to folate. For the study of histone modifications, we adopted a

hypothesis driven approach and targeted assays to key cytokine and transcription factor

loci known to be involved in the development of allergic disease. This was in part due

to the technologies available, logistics and expenses.

97

In chapter 3, we performed a comparison of DNA methylation profiles from two

neonatally derived leukocyte populations according to the level of in utero folate

exposure. Our research efforts were directed by increasing concerns over inadvertent

adverse effects of potentially excessive maternal folate intake on offspring long-term

health outcomes and evidence from animal studies suggesting DNA methylation is the

likely mechanism of such effects. It was necessary to test this in human cohorts as

animal studies, although more tractable, may not precisely reflect the events occurring

in human epigenome.

Analysis of the microarray data revealed that DNA methylation levels at long

interspersed nucleotide element-1 (LINE-1) repeat elements and average level of DNA

methylation in the genome were not different according folate status in both CD4+ T-

cells and APC. This broad view of the neonatal methylome suggests that the neonatal

methylome is in general, resistant to changes in utero folate exposure.

Despite the global methylation status being largely unperturbed, we detected several

DMR associated with folate status, the most striking of which was a 923 bp region

broadly hypomethylated in neonates exposed to higher levels of folate in utero.

Interestingly this de-methylated region was located in a CpG island upstream of the

promoter region of ZFP57, a gene with a known role in the regulation of DNA

methylation. Analysis of H3 and H4 histone acetylation in the promoter region of

ZFP57 revealed relatively higher acetylation levels in neonates from HF group

indicating that hypomethylation of the upstream region is associated with a more

transcriptionally permissive histone mark in this group. ZFP57 mRNA expression was

higher in samples from the HF group and this is consistent with a reduction in DNA

methylation in the upstream region. We validated this key finding in an independent set

of samples using an alternative technology, Sequenom EpiTyper. Both the microarray

screen and the EpiTyper data were concordant for de-methylation in this region in both

CD4+ T-cell and APC cells.

ZFP57 plays a central role in regulating imprinting marks during development through

preferentially binding to methylated DNA in imprinted domains. This binding leads to

recruitment of DNMT to target sites to initiate or maintain the methylation of the

genomic region rendering the region transcriptionally inactive (146, 147). Aberrant

expression of imprinted genes are associated with a number of developmental disorders

98

(302), thus maintaining stable expression pattern of this region is crucial for the

development. Our genome-wide screening did not observe difference in DNA

methylation levels at imprinted control regions in high vs low folate groups indicating a

compensatory mechanism or a negative feed-back loop may exist to regulate the activity

of the ZFP57 gene in response to folate exposure to stabilise DNA methylation levels at

imprinting control regions. It is also plausible that the magnitude of difference in the

ZFP57 transcript was not sufficient to induce changes in imprinted genes or the level of

ZFP57 protein was not changed sufficiently to alter the methylation of other genes.

Analysis of dose-response relationship of folic acid on the activity of ZFP57 in

maintaining DNA methylation at imprinting control regions in mouse models that are

knocked out for the upstream regulatory region would be useful to study the precise role

of this region and is an area for future research.

Emerging evidence suggests that ZFP57 may have other functions beyond imprinting

establishment and maintenance; ZFP57 is expressed in low but detectable levels in adult

tissues including peripheral blood mononuclear cells (303), ZFP57 mediates up-

regulation in anchorage-independent growth of cancer cells in mice pointing to

oncogenic properties of ZFP57 (304). Interestingly, a recent study by Andrew

Feinberg’s group found up-stream region of ZFP57, a similar region to that we have

reported here, is differentially methylated in post mortem brain tissues from individuals

with autism spectrum disorders compared to brain tissue of healthy individuals (305).

This together with our finding indicates that aberrant methylation in this nutritionally

sensitive genomic region may be involved in disease pathogenesis and is a logical

candidate for studies investigating DOHaD.

Data from histone acetylation assays performed on the same sample population (chapter

4) revealed higher histone acetylation levels at GATA3 and IL9 loci in neonates exposed

to higher folate levels. GATA-3, the master regulator of Th2 cell lineage, activates

transcription of Th2 cytokine loci and stabilizes the differentiated state by repressing

activation of the alternative Th1 cell fate (306). Initially described as a Th2 cytokine,

IL-9 has now shown to be produced by other Th subsets and is associated with allergic

inflammation and autoimmune diseases (307, 308). These histone modifications did not

appear to alter the gene activity as determined by analysis of mRNA expression levels

in both groups. Furthermore we did not observer any concurrent changes in DNA

methylation using the microarray screen. Therefore, the biological significance of these

99

differentially acetylated histones is unclear. It is possible that they represent loci poised

for increased transcriptional activity in response to activation-induced signals. Future

studies should aim to study the activation response in addition to baseline histone

acetylation and DNA methylation levels.

Our data provide direct evidence that maternal folic acid consumption has the capacity

to modify developmental DNA methylation profiles at site-specific loci. This is the first

genome-wide study to describe these effects in humans and contribute to a growing

body of data regarding epigenetic programing of in utero nutritional exposures. Despite

these effects described here, contribution of higher levels of maternal folate to increased

risk of allergic diseases is not clear. Based on the lack of strong evidence for functional

epigenetic changes at allergy-associated loci in the experiments described here, it is

unlikely that high folic acid levels modify the risk for disease via this mechanism which

is in contrast to the findings of the highly cited mouse study by Hollingsworth et al.

Rather, it is plausible that ZFP57 may play a role in compensating the effects of folate

exposure by maintaining a stable DNA methylation level at these loci. Would this

mechanism be biologically plausible and within what range of folate exposure it would

occur are questions that need to be addressed in future studies.

In chapter 5, we performed a between-group comparison of genome-wide DNA

methylation profiles from a randomised controlled trial of maternal fish oil

supplementation during pregnancy. We studied global methylation profiles from CD4+

T-cells in neonates exposed or not exposed to n-3 PUFA in utero. Our main hypothesis

was that the beneficial effects of maternal fish oil intake during pregnancy are mediated

through epigenetic modifications. However, analysis of DNA methylation microarray

data did not provide clear evidence of substantial epigenetic effects of maternal fish oil

intake on the neonate. There were no CpG sites that were substantially modified

between treatment groups, detected effects were small and unlikely to be of biological

significance. These observations were made in a well-designed study cohort, in which

priori evidence of favourable immune outcomes of maternal fish oil supplementation

has been reported. Based on the available data, there was no convincing evidence that

biological pathways involved in mediating anti-inflammatory effects of n-3 PUFA are

likely to be regulated by DNA methylation, at least in CD4+ T-cells. It is possible that

neonatal erythrocytes, in which direct effects of the supplementation on lipid profile

were observed, may harbour CpG sites or regions amenable for modulation by maternal

100

fish oil, affecting downstream biological processes. Nevertheless, such modulations in

red cells, if present, are unlikely to have direct effects on immune development.

Folate is a crucial component of the one-carbon mediated pathway, therefore, is a

potential candidate for epigenetic regulation whereas the links with fish oil and DNA

methylation are less clear. It remains possible that the well described biological effects

of n-3 PUFA are more likely to be mediated by other epigenetic or post-transcriptional

effects that modulate cellular function. In animals, feeding dams with high fat diets was

associated with alterations in histone marks (309, 310) and reduced expression of

ncRNA (310) in the offspring supporting this notion. Studies are currently underway by

our group to extend the findings of animal experiments to human cohorts.

Collectively, the data suggest that some maternal nutritional factors have the capacity to

modulate the fetal epigenome, of which with direct effects on one-carbon mediated

pathway have the potential to exert biologically significant effects through direct effects

on DNA methylation.

6.2 Limitations of the study

The field of epigenetic epidemiology is a relatively new area of research and its

application to DOHaD research has not been extensively elaborated. While there has

been rapid advancement in technology to characterise and analyse epigenomic data over

the past 4-5 years, further evidence is required to conceptualise the study design to

achieve the maximum research output in a cost-effective manner.

In human populations the most powerful study design is the randomised controlled trial

(RCT), which can provide level one evidence regarding the effect of specific exposures.

In chapter 5 we were able to utilise such a study design, however, in chapter 4 this was

not possible given the ethical limitations of a RCT of folic acid during pregnancy.

Therefore, we were restricted to sampling from a normally distributed population. For

the discovery phase of this study we sampled individuals from the extremes of the

distribution curve in a classical ‘extremes of exposure’ design. In comparison to random

sampling, selecting individuals from the phenotypic extremes of a binary trait has

shown to increase the statistical power to discover genetic variation contributing to the

trait in GWAS (311-313). Given the high cost of genomics experiments, this type of

design maximises power to detect biological effects in a small population, however,

101

replication of findings becomes necessary due to the potential to sample individuals

who may differ in some way by chance alone. The disadvantages of such an approach

are that the results may not represent the true effect size in the total population and the

analysis is limited to the selected phenotype (313, 314). Regardless, this approach is

useful to identify relevant candidates in small cohort which then can be assessed in a

large population using a more cost-effective targeted approach. The data generated here

by this approach can now form the basis of large scale follow-up studies.

The methylation studies of this thesis were of exploratory in nature enabling

identification of DMR in response to in utero nutritional exposures. We were restricted

to employ a selected candidate approach to study histone modifications due to

requirement of higher starting material for genome-wide histone assay and to limited

available funds and it is possible that loci other than reported here may harbour histones

potentially modulated by maternal folate. We did not observe any difference in

stimulated-CBMC cytokine profile between folate groups. However, it is likely that

significant changes in the cytokine profile may not be apparent with the small number

of individuals examined for each group (11 vs 12).

Analysis of peripheral blood is considered appropriate for investigating epigenetic

modulation in immune-related diseases/phenotypes, however, only relevant tissue

samples would inform the site-specific epigenetic changes occurred following an

environmental exposure. Cord blood is one of the relatively easily accessible neonatal

biological samples, therefore, in this thesis cord blood derived leukocytes were analysed

as the reference cellular material to study the epigenetic immune programing of the

neonate. Analysis of multiple neonatal tissues would provide the opportunity to study

the degree to which different tissues show varying sensitivities to the same maternal

exposures and the extent to which this interaction influence the epigenetic programing

and disease risk, however, may not be practical in human studies. Also, it remains

unclear whether postnatal accumulation of environmentally mediated epigenetic

changes have the potential to ‘overwrite’ the epigenetic programming established at

birth. The epigenetic ‘blue print’ in cord blood derived immune cells may undergo

developmental changes in the first years of life which also pose a window of

opportunity for immune development and maturation that influence the later life health

and disease risk. Longitudinal birth cohorts with detailed collection of environmental

and other data and epigenetic analysis of multiple biospecimens will greatly facilitate

102

our understanding of nutritionally mediated epigenetic programing in influencing the

disease risk.

Despite these limitations this thesis has identified a number of neonatal epigenetic

marks that are amenable for modifications by maternal nutritional exposures upon

which detailed investigations can be carried out in future studies.

6.3 Directions for future studies in this area

The results obtained through the present studies make a substantial contribution to

advance our knowledge of nutritional programming of the fetal epigenome. Our data

provides direct evidence in humans that these effects occur but also highlight the

complexity of epigenetic dynamics. They provide direction for further studies in this

evolving area of nutri-epigenetics and recommendations for future studies are given

below.

Epigenetic regulation of gene expression occurs at multiple levels, therefore,

investigations should not be limited to DNA methylation and histone modifications.

Future studies in this area should consider investigating the contribution from other

epigenetic mechanisms such as miRNA and chromatin remodelling complexes possibly

on a genome-wide scale. It is necessary to develop cross-platform workflows for this

purposes, however, integration of multiple genome-wide molecular data sets pose huge

challenges during data processing, statistical analyses and interpreting the data.

Limited access to relevant tissue samples is an issue in human epigenetic studies, more

in studies in fetal programming. Non-invasive techniques (i.e. imaging techniques) of

epigenetic profiling will revolutionise the field of epigenetic studies enabling profiling

epigenetic marks across multiple fetal (and maternal) tissues simultaneously. Such

technology will also enable tracking dynamic epigenetic changes at different time points

during pregnancy that facilitates quantification of individual epigenetic variation to a

specific exposure.

Recent data from animal experiments points towards a role of maternal diet in shaping

the offspring gut microbiome (315-318). In mice, feeding dams a diet rich in n-3 fatty

acids have resulted in altered offspring gut microbiome, anti-inflammatory immune

response and reduced allergic reactivity to allergens suggesting that immune modulating

103

effects of maternal diet may mediate through gut microbiome. While, evidence from

animal studies are suggestive, it is necessary to replicate these findings in humans as

such randomised clinical trials of maternal supplementations with relevant

biospecimens would provide the opportunity to test this hypothesis. A recent landmark

study by Aagaard and colleagues revealed that the placenta is not devoid of microflora

as we previously thought and the pattern of placental microbiome resembles tissue-

specific maternal microbiome (319). While, it requires more studies in support of this

observation before any conclusions are made, it is intriguing to speculate that a

microbiome at the feto-maternal interface could play an important role in shaping the

fetal immune development. Furthermore, maternal dietary factors such as fish oil

supplementation may have effects on the placental microbiome with consequences on

fetal immune programing and should be investigated in future studies.

6.4 Concluding remarks

The dramatic rise in a whole range of NCD including cardiovascular, metabolic and

allergic diseases is a major health crisis of the modern world. Developmental Origin of

Health and Disease hypothesis proposed that these conditions have their origin in fetal

life and the events occurring during pregnancy play a crucial role in determining the

disease risk. The programming effects of maternal nutrition has been clearly linked with

infant immune diseases highlighting the importance of maternal nutrition in influencing

immune development and maturation. There is hope that intense research interest in

elaborating mechanistic links of dysregulated immune development following early

exposure to a number of environmental factors will shed light on better understanding

disease development and on future intervention and/or prevention strategies. In this

context, studies investigating epigenetic mechanisms as the likely mediator of

environment induced-aberrant immune development are likely to provide valuable

information on understanding disease pathogenesis.

Using cutting-edge epigenomic profiling techniques, this thesis has provided evidence

for maternal nutritional factors can modulate fetal developmental epigenome that may

have clear implications for developmental health. It further highlights the complex

interplay between different layers of epigenetic processes in regulating downstream

effects of environmental exposures and should be an important consideration in all

future studies of this nature.

104

This thesis has identified several epigenomic regions that are susceptible to modulation

by maternal nutritional factors, providing a frame work for more detailed investigations

towards identifying strategies to promote optimum immune development that will

ultimately aid reducing the burden of inflammatory NCD.

105

BIBLIOGRAPHY 1. Bach JF. The effect of infections on susceptibility to autoimmune and allergic

diseases. N Engl J Med. 2002;347(12):911-20.

2. Prescott SL. Early-life environmental determinants of allergic diseases and the

wider pandemic of inflammatory noncommunicable diseases. J Allergy Clin

Immunol. 2013;131(1):23-30.

3. Kolb H, Mandrup-Poulsen T. The global diabetes epidemic as a consequence of

lifestyle-induced low-grade inflammation. Diabetologia. 2010;53(1):10-20.

4. Prescott SL. Early origins of allergic disease: a review of processes and influences

during early immune development. Curr Opin Allergy Clin Immunol.

2003;3(2):125-32.

5. Saito S. Cytokine network at the feto-maternal interface. J Reprod Immunol.

2000;47(2):87-103.

6. Breckler LA, Hale J, Jung W, Westcott L, Dunstan JA, Thornton CA, et al.

Modulation of in vivo and in vitro cytokine production over the course of

pregnancy in allergic and non-allergic mothers. Pediatr Allergy Immunol.

2010;21(1 Pt 1):14-21.

7. Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine

interactions in the maternal-fetal relationship: is successful pregnancy a TH2

phenomenon? Immunol Today. 1993;14(7):353-6.

8. Piccinni MP, Scaletti C, Maggi E, Romagnani S. Role of hormone-controlled

Th1- and Th2-type cytokines in successful pregnancy. J Neuroimmunol.

2000;109(1):30-3.

9. Schumacher A, Brachwitz N, Sohr S, Engeland K, Langwisch S, Dolaptchieva M,

et al. Human chorionic gonadotropin attracts regulatory T cells into the fetal-

maternal interface during early human pregnancy. J Immunol. 2009;182(9):5488-

97.

10. Prescott SL, Tulic M, Kumah AO, Richman T, Crook M, Martino D, et al.

Reduced placental FOXP3 associated with subsequent infant allergic disease. J

Allergy Clin Immunol. 2011;128(4):886-7 e5.

11. Lewis RM, Demmelmair H, Gaillard R, Godfrey KM, Hauguel-de Mouzon S,

Huppertz B, et al. The placental exposome: placental determinants of fetal

adiposity and postnatal body composition. Annals of nutrition & metabolism.

2013;63(3):208-15.

106

12. Novakovic B, Saffery R. The ever growing complexity of placental epigenetics -

role in adverse pregnancy outcomes and fetal programming. Placenta.

2012;33(12):959-70.

13. Koning H, Baert MR, Oranje AP, Savelkoul HF, Neijens HJ. Development of

immune functions related to allergic mechanisms in young children. Pediatr Res.

1996;40(3):363-75.

14. Mold JE, McCune JM. At the crossroads between tolerance and aggression:

Revisiting the "layered immune system" hypothesis. Chimerism. 2011;2(2):35-41.

15. Le Douarin NM, Jotereau FV. Tracing of cells of the avian thymus through

embryonic life in interspecific chimeras. J Exp Med. 1975;142(1):17-40.

16. Havran WL, Allison JP. Developmentally ordered appearance of thymocytes

expressing different T-cell antigen receptors. Nature. 1988;335(6189):443-5.

17. Havran WL, Allison JP. Origin of Thy-1+ dendritic epidermal cells of adult mice

from fetal thymic precursors. Nature. 1990;344(6261):68-70.

18. Ikuta K, Kina T, MacNeil I, Uchida N, Peault B, Chien YH, et al. A

developmental switch in thymic lymphocyte maturation potential occurs at the

level of hematopoietic stem cells. Cell. 1990;62(5):863-74.

19. Mold JE, Venkatasubrahmanyam S, Burt TD, Michaelsson J, Rivera JM, Galkina

SA, et al. Fetal and adult hematopoietic stem cells give rise to distinct T cell

lineages in humans. Science. 2010;330(6011):1695-9.

20. Claas FH, Gijbels Y, van der Velden-de Munck J, van Rood JJ. Induction of B

cell unresponsiveness to noninherited maternal HLA antigens during fetal life.

Science. 1988;241(4874):1815-7.

21. Cupedo T, Nagasawa M, Weijer K, Blom B, Spits H. Development and activation

of regulatory T cells in the human fetus. Eur J Immunol. 2005;35(2):383-90.

22. Upham JW, Lee PT, Holt BJ, Heaton T, Prescott SL, Sharp MJ, et al.

Development of interleukin-12-producing capacity throughout childhood. Infect

Immun. 2002;70(12):6583-8.

23. Langrish CL, Buddle JC, Thrasher AJ, Goldblatt D. Neonatal dendritic cells are

intrinsically biased against Th-1 immune responses. Clin Exp Immunol.

2002;128(1):118-23.

24. Prescott SL, Macaubas C, Smallacombe T, Holt BJ, Sly PD, Holt PG.

Development of allergen-specific T-cell memory in atopic and normal children.

Lancet. 1999;353(9148):196-200.

107

25. Chen L, Cohen AC, Lewis DB. Impaired allogeneic activation and T-helper 1

differentiation of human cord blood naive CD4 T cells. Biology of blood and

marrow transplantation : journal of the American Society for Blood and Marrow

Transplantation. 2006;12(2):160-71.

26. Tulic MK, Andrews D, Crook ML, Charles A, Tourigny MR, Moqbel R, et al.

Changes in thymic regulatory T-cell maturation from birth to puberty: differences

in atopic children. J Allergy Clin Immunol. 2012;129(1):199-206 e1-4.

27. Tulic MK, Hodder M, Forsberg A, McCarthy S, Richman T, D'Vaz N, et al.

Differences in innate immune function between allergic and nonallergic children:

new insights into immune ontogeny. J Allergy Clin Immunol. 2011;127(2):470-8

e1.

28. Prescott SL, Noakes P, Chow BW, Breckler L, Thornton CA, Hollams EM, et al.

Presymptomatic differences in Toll-like receptor function in infants who have

allergy. J Allergy Clin Immunol. 2008;122(2):391-9, 9 e1-5.

29. Prescott SL, Taylor A, King B, Dunstan J, Upham JW, Thornton CA, et al.

Neonatal interleukin-12 capacity is associated with variations in allergen-specific

immune responses in the neonatal and postnatal periods. Clin Exp Allergy.

2003;33(5):566-72.

30. Smith M, Tourigny MR, Noakes P, Thornton CA, Tulic MK, Prescott SL.

Children with egg allergy have evidence of reduced neonatal

CD4(+)CD25(+)CD127(lo/-) regulatory T cell function. J Allergy Clin Immunol.

2008;121(6):1460-6, 6 e1-7.

31. Devereux G, Barker RN, Seaton A. Antenatal determinants of neonatal immune

responses to allergens. Clin Exp Allergy. 2002;32(1):43-50.

32. Pfefferle PI, Prescott SL, Kopp M. Microbial influence on tolerance and

opportunities for intervention with prebiotics/probiotics and bacterial lysates. J

Allergy Clin Immunol. 2013;131(6):1453-63; quiz 64.

33. Martino D, Holt P, Prescott S. A novel role for interleukin-1 receptor signaling in

the developmental regulation of immune responses to endotoxin. Pediatr Allergy

Immunol. 2012;23(6):567-72.

34. Yerkovich ST, Wikstrom ME, Suriyaarachchi D, Prescott SL, Upham JW, Holt

PG. Postnatal development of monocyte cytokine responses to bacterial

lipopolysaccharide. Pediatr Res. 2007;62(5):547-52.

35. Fujimaki W, Takahashi N, Ohnuma K, Nagatsu M, Kurosawa H, Yoshida S, et al.

Comparative study of regulatory T cell function of human CD25CD4 T cells from

108

thymocytes, cord blood, and adult peripheral blood. Clinical & developmental

immunology. 2008;2008:305859.

36. Delespesse G, Yang LP, Ohshima Y, Demeure C, Shu U, Byun DG, et al.

Maturation of human neonatal CD4+ and CD8+ T lymphocytes into Th1/Th2

effectors. Vaccine. 1998;16(14-15):1415-9.

37. Maeda Y, Noda S, Tanaka K, Sawamura S, Aiba Y, Ishikawa H, et al. The failure

of oral tolerance induction is functionally coupled to the absence of T cells in

Peyer's patches under germfree conditions. Immunobiology. 2001;204(4):442-57.

38. Nylund L, Satokari R, Nikkila J, Rajilic-Stojanovic M, Kalliomaki M, Isolauri E,

et al. Microarray analysis reveals marked intestinal microbiota aberrancy in

infants having eczema compared to healthy children in at-risk for atopic disease.

BMC Microbiol. 2013;13:12.

39. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic

Estonian and Swedish 2-year-old children. Clin Exp Allergy. 1999;29(3):342-6.

40. West CE, Videky DJ, Prescott SL. Role of diet in the development of immune

tolerance in the context of allergic disease. Curr Opin Pediatr. 2010;22(5):635-41.

41. Castro-Rodriguez JA, Garcia-Marcos L, Alfonseda Rojas JD, Valverde-Molina J,

Sanchez-Solis M. Mediterranean diet as a protective factor for wheezing in

preschool children. J Pediatr. 2008;152(6):823-8, 8 e1-2.

42. Chatzi L, Torrent M, Romieu I, Garcia-Esteban R, Ferrer C, Vioque J, et al.

Mediterranean diet in pregnancy is protective for wheeze and atopy in childhood.

Thorax. 2008;63(6):507-13.

43. Viscogliosi G, Cipriani E, Liguori ML, Marigliano B, Saliola M, Ettorre E, et al.

Mediterranean Dietary Pattern Adherence: Associations with Prediabetes,

Metabolic Syndrome, and Related Microinflammation. Metab Syndr Relat Disord.

2013.

44. Sofi F, Abbate R, Gensini GF, Casini A. Accruing evidence on benefits of

adherence to the Mediterranean diet on health: an updated systematic review and

meta-analysis. Am J Clin Nutr. 2010;92(5):1189-96.

45. Matzinger P, Kamala T. Tissue-based class control: the other side of tolerance.

Nat Rev Immunol. 2011;11(3):221-30.

46. Prescott SL, Dunstan JA. Prenatal fatty acid status and immune development: the

pathways and the evidence. Lipids. 2007;42(9):801-10.

109

47. Seifried HE, Anderson DE, Fisher EI, Milner JA. A review of the interaction

among dietary antioxidants and reactive oxygen species. J Nutr Biochem.

2007;18(9):567-79.

48. Stulnig TM, Zeyda M. Immunomodulation by polyunsaturated fatty acids: impact

on T-cell signaling. Lipids. 2004;39(12):1171-5.

49. Bagga D, Wang L, Farias-Eisner R, Glaspy JA, Reddy ST. Differential effects of

prostaglandin derived from omega-6 and omega-3 polyunsaturated fatty acids on

COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A.

2003;100(4):1751-6.

50. Ariel A, Serhan CN. Resolvins and protectins in the termination program of acute

inflammation. Trends Immunol. 2007;28(4):176-83.

51. Weise C, Heunemann C, Loddenkemper C, Herz U, van Tol EA, Worm M.

Dietary docosahexaenoic acid in combination with arachidonic acid ameliorates

allergen-induced dermatitis in mice. Pediatr Allergy Immunol. 2011;22(5):497-

504.

52. Barden AE, Mori TA, Dunstan JA, Taylor AL, Thornton CA, Croft KD, et al.

Fish oil supplementation in pregnancy lowers F2-isoprostanes in neonates at high

risk of atopy. Free Radic Res. 2004;38(3):233-9.

53. Prescott SL, Barden AE, Mori TA, Dunstan JA. Maternal fish oil supplementation

in pregnancy modifies neonatal leukotriene production by cord-blood-derived

neutrophils. Clin Sci (Lond). 2007;113(10):409-16.

54. Dunstan JA, Mori TA, Barden A, Beilin LJ, Taylor AL, Holt PG, et al. Fish oil

supplementation in pregnancy modifies neonatal allergen-specific immune

responses and clinical outcomes in infants at high risk of atopy: a randomized,

controlled trial. J Allergy Clin Immunol. 2003;112(6):1178-84.

55. Palmer DJ, Sullivan T, Gold MS, Prescott SL, Heddle R, Gibson RA, et al. Effect

of n-3 long chain polyunsaturated fatty acid supplementation in pregnancy on

infants' allergies in first year of life: randomised controlled trial. BMJ.

2012;344:e184.

56. Furuhjelm C, Warstedt K, Larsson J, Fredriksson M, Bottcher MF, Falth-

Magnusson K, et al. Fish oil supplementation in pregnancy and lactation may

decrease the risk of infant allergy. Acta Paediatr. 2009;98(9):1461-7.

57. D'Vaz N, Meldrum SJ, Dunstan JA, Martino D, McCarthy S, Metcalfe J, et al.

Postnatal fish oil supplementation in high-risk infants to prevent allergy:

randomized controlled trial. Pediatrics. 2012;130(4):674-82.

110

58. Sausenthaler S, Koletzko S, Schaaf B, Lehmann I, Borte M, Herbarth O, et al.

Maternal diet during pregnancy in relation to eczema and allergic sensitization in

the offspring at 2 y of age. Am J Clin Nutr. 2007;85(2):530-7.

59. Oien T, Storro O, Johnsen R. Do early intake of fish and fish oil protect against

eczema and doctor-diagnosed asthma at 2 years of age? A cohort study. J

Epidemiol Community Health. 2010;64(2):124-9.

60. Hodge L, Salome CM, Peat JK, Haby MM, Xuan W, Woolcock AJ. Consumption

of oily fish and childhood asthma risk. Med J Aust. 1996;164(3):137-40.

61. Utsugi M, Dobashi K, Ishizuka T, Endou K, Hamuro J, Murata Y, et al. c-Jun N-

terminal kinase negatively regulates lipopolysaccharide-induced IL-12 production

in human macrophages: role of mitogen-activated protein kinase in glutathione

redox regulation of IL-12 production. J Immunol. 2003;171(2):628-35.

62. Devereux G, Turner SW, Craig LC, McNeill G, Martindale S, Harbour PJ, et al.

Low maternal vitamin E intake during pregnancy is associated with asthma in 5-

year-old children. Am J Respir Crit Care Med. 2006;174(5):499-507.

63. Martindale S, McNeill G, Devereux G, Campbell D, Russell G, Seaton A.

Antioxidant intake in pregnancy in relation to wheeze and eczema in the first two

years of life. Am J Respir Crit Care Med. 2005;171(2):121-8.

64. Forastiere F, Pistelli R, Sestini P, Fortes C, Renzoni E, Rusconi F, et al.

Consumption of fresh fruit rich in vitamin C and wheezing symptoms in children.

SIDRIA Collaborative Group, Italy (Italian Studies on Respiratory Disorders in

Children and the Environment). Thorax. 2000;55(4):283-8.

65. Okoko B, Burney P, Newson R, Potts J, Shaheen S. Childhood asthma and fruit

consumption in South London. The European Respiratory Journal. 2007:1-24.

66. Nurmatov U, Devereux G, Sheikh A. Nutrients and foods for the primary

prevention of asthma and allergy: systematic review and meta-analysis. J Allergy

Clin Immunol. 2011;127(3):724-33 e1-30.

67. Murr C, Schroecksnadel K, Winkler C, Ledochowski M, Fuchs D. Antioxidants

may increase the probability of developing allergic diseases and asthma. Med

Hypotheses. 2005;64(5):973-7.

68. Alm JS, Swartz J, Lilja G, Scheynius A, Pershagen G. Atopy in children of

families with an anthroposophic lifestyle. Lancet. 1999;353(9163):1485-8.

69. Wickens K, Pearce N, Crane J, Beasley R. Antibiotic use in early childhood and

the development of asthma. Clin Exp Allergy. 1999;29(6):766-71.

111

70. Strachan DP. Hay fever, hygiene, and household size. BMJ.

1989;299(6710):1259-60.

71. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the

gut microbiome and the immune system. Nature. 2011;474(7351):327-36.

72. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic

endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761-

72.

73. Shi L, Li M, Miyazawa K, Li Y, Hiramatsu M, Xu J, et al. Effects of heat-

inactivated Lactobacillus gasseri TMC0356 on metabolic characteristics and

immunity of rats with the metabolic syndrome. Br J Nutr. 2013;109(2):263-72.

74. Gaboriau-Routhiau V, Raibaud P, Dubuquoy C, Moreau MC. Colonization of

gnotobiotic mice with human gut microflora at birth protects against Escherichia

coli heat-labile enterotoxin-mediated abrogation of oral tolerance. Pediatr Res.

2003;54(5):739-46.

75. Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of

intestinal bacterial flora for the development of an IgE production system fully

susceptible to oral tolerance induction. J Immunol. 1997;159(4):1739-45.

76. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of

inflammatory responses by gut microbiota and chemoattractant receptor GPR43.

Nature. 2009;461(7268):1282-6.

77. Arslanoglu S, Moro GE, Schmitt J, Tandoi L, Rizzardi S, Boehm G. Early dietary

intervention with a mixture of prebiotic oligosaccharides reduces the incidence of

allergic manifestations and infections during the first two years of life. J Nutr.

2008;138(6):1091-5.

78. Gruber C, van Stuijvenberg M, Mosca F, Moro G, Chirico G, Braegger CP, et al.

Reduced occurrence of early atopic dermatitis because of immunoactive prebiotics

among low-atopy-risk infants. J Allergy Clin Immunol. 2010;126(4):791-7.

79. Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G. A mixture of

prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the

first six months of age. Arch Dis Child. 2006;91(10):814-9.

80. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An

obesity-associated gut microbiome with increased capacity for energy harvest.

Nature. 2006;444(7122):1027-31.

112

81. Holmes E, Li JV, Marchesi JR, Nicholson JK. Gut microbiota composition and

activity in relation to host metabolic phenotype and disease risk. Cell Metab.

2012;16(5):559-64.

82. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut

microbiota as an environmental factor that regulates fat storage. Proc Natl Acad

Sci U S A. 2004;101(44):15718-23.

83. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al.

A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480-4.

84. Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357(3):266-81.

85. Lucas A. Programming by early nutrition: an experimental approach. J Nutr.

1998;128(2 Suppl):401S-6S.

86. Barker DJ, Fall CH. Fetal and infant origins of cardiovascular disease. Arch Dis

Child. 1993;68(6):797-9.

87. Barker DJ. In utero programming of chronic disease. Clin Sci (Lond).

1998;95(2):115-28.

88. Gluckman PD, Hanson MA. Developmental origins of disease paradigm: a

mechanistic and evolutionary perspective. Pediatr Res. 2004;56(3):311-7.

89. Martino DJ, Prescott SL. Silent mysteries: epigenetic paradigms could hold the

key to conquering the epidemic of allergy and immune disease. Allergy.

2010;65(1):7-15.

90. Desai M, Crowther NJ, Lucas A, Hales CN. Organ-selective growth in the

offspring of protein-restricted mothers. Br J Nutr. 1996;76(4):591-603.

91. Lucas A, Baker BA, Desai M, Hales CN. Nutrition in pregnant or lactating rats

programs lipid metabolism in the offspring. Br J Nutr. 1996;76(4):605-12.

92. Sinclair KD, Allegrucci C, Singh R, Gardner DS, Sebastian S, Bispham J, et al.

DNA methylation, insulin resistance, and blood pressure in offspring determined

by maternal periconceptional B vitamin and methionine status. Proc Natl Acad Sci

U S A. 2007;104(49):19351-6.

93. Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, et al. In

utero supplementation with methyl donors enhances allergic airway disease in

mice. J Clin Invest. 2008;118(10):3462-9.

94. Gluckman PD, Hanson MA, Spencer HG. Predictive adaptive responses and

human evolution. Trends Ecol Evol. 2005;20(10):527-33.

95. Lee TM, Zucker I. Vole infant development is influenced perinatally by maternal

photoperiodic history. Am J Physiol. 1988;255(5 Pt 2):R831-8.

113

96. McDade TW. Early environments and the ecology of inflammation. Proc Natl

Acad Sci U S A. 2012;109 Suppl 2:17281-8.

97. Rinas U, Horneff G, Wahn V. Interferon-gamma production by cord-blood

mononuclear cells is reduced in newborns with a family history of atopic disease

and is independent from cord blood IgE-levels. Pediatr Allergy Immunol.

1993;4(2):60-4.

98. Tang ML, Kemp AS, Thorburn J, Hill DJ. Reduced interferon-gamma secretion in

neonates and subsequent atopy. Lancet. 1994;344(8928):983-5.

99. D'Vaz N, Ma Y, Dunstan JA, Lee-Pullen TF, Hii C, Meldrum S, et al. Neonatal

protein kinase C zeta expression determines the neonatal T-Cell cytokine

phenotype and predicts the development and severity of infant allergic disease.

Allergy. 2012.

100. Martino DJ, Bosco A, McKenna KL, Hollams E, Mok D, Holt PG, et al. T-cell

activation genes differentially expressed at birth in CD4+ T-cells from children

who develop IgE food allergy. Allergy. 2012;67(2):191-200.

101. Braun-Fahrlander C, Riedler J, Herz U, Eder W, Waser M, Grize L, et al.

Environmental exposure to endotoxin and its relation to asthma in school-age

children. N Engl J Med. 2002;347(12):869-77.

102. Nowak D, Heinrich J, Jorres R, Wassmer G, Berger J, Beck E, et al. Prevalence of

respiratory symptoms, bronchial hyperresponsiveness and atopy among adults:

west and east Germany. Eur Respir J. 1996;9(12):2541-52.

103. Ponsonby AL, Dwyer T, Kemp A, Couper D, Cochrane J, Carmichael A. A

prospective study of the association between home gas appliance use during

infancy and subsequent dust mite sensitization and lung function in childhood.

Clin Exp Allergy. 2001;31(10):1544-52.

104. Ottman R. Gene-environment interaction: definitions and study designs.

Preventive medicine. 1996;25(6):764-70.

105. Maher B. Personal genomes: The case of the missing heritability. Nature.

2008;456(7218):18-21.

106. McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JP, et

al. Genome-wide association studies for complex traits: consensus, uncertainty

and challenges. Nat Rev Genet. 2008;9(5):356-69.

107. Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al.

Finding the missing heritability of complex diseases. Nature.

2009;461(7265):747-53.

114

108. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental

origins hypothesis. Annu Rev Nutr. 2007;27:363-88.

109. Gluckman PD, Hanson MA, Mitchell MD. Developmental origins of health and

disease: reducing the burden of chronic disease in the next generation. Genome

Med. 2010;2(2):14.

110. Kota SK, Feil R. Epigenetic transitions in germ cell development and meiosis.

Developmental cell. 2010;19(5):675-86.

111. Johannes F, Porcher E, Teixeira FK, Saliba-Colombani V, Simon M, Agier N, et

al. Assessing the impact of transgenerational epigenetic variation on complex

traits. PLoS Genet. 2009;5(6):e1000530.

112. Daxinger L, Whitelaw E. Understanding transgenerational epigenetic inheritance

via the gametes in mammals. Nat Rev Genet. 2012;13(3):153-62.

113. Ballestar E, Wolffe AP. Methyl-CpG-binding proteins. Targeting specific gene

repression. Eur J Biochem. 2001;268(1):1-6.

114. Severin PM, Zou X, Gaub HE, Schulten K. Cytosine methylation alters DNA

mechanical properties. Nucleic Acids Res. 2011;39(20):8740-51.

115. Holliday R, Grigg GW. DNA methylation and mutation. Mutat Res.

1993;285(1):61-7.

116. Li Y, Zhu J, Tian G, Li N, Li Q, Ye M, et al. The DNA methylome of human

peripheral blood mononuclear cells. PLoS Biol. 2010;8(11):e1000533.

117. Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, Hon G, et al. Hotspots of

aberrant epigenomic reprogramming in human induced pluripotent stem cells.

Nature. 2011;471(7336):68-73.

118. Ramsahoye BH, Biniszkiewicz D, Lyko F, Clark V, Bird AP, Jaenisch R. Non-

CpG methylation is prevalent in embryonic stem cells and may be mediated by

DNA methyltransferase 3a. Proc Natl Acad Sci U S A. 2000;97(10):5237-42.

119. Chen T, Li E. Establishment and maintenance of DNA methylation patterns in

mammals. Curr Top Microbiol Immunol. 2006;301:179-201.

120. Cantone I, Fisher AG. Epigenetic programming and reprogramming during

development. Nat Struct Mol Biol. 2013;20(3):282-9.

121. Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell.

2007;128(4):635-8.

122. Williams K, Christensen J, Helin K. DNA methylation: TET proteins-guardians of

CpG islands? EMBO Rep. 2012;13(1):28-35.

115

123. Ito S, D'Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet

proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass

specification. Nature. 2010;466(7310):1129-33.

124. Talbert PB, Henikoff S. Histone variants--ancient wrap artists of the epigenome.

Nat Rev Mol Cell Biol. 2010;11(4):264-75.

125. Hargreaves DC, Crabtree GR. ATP-dependent chromatin remodeling: genetics,

genomics and mechanisms. Cell Res. 2011;21(3):396-420.

126. Berger SL. The complex language of chromatin regulation during transcription.

Nature. 2007;447(7143):407-12.

127. Dion MF, Altschuler SJ, Wu LF, Rando OJ. Genomic characterization reveals a

simple histone H4 acetylation code. Proc Natl Acad Sci U S A.

2005;102(15):5501-6.

128. Sanchez R, Zhou MM. The role of human bromodomains in chromatin biology

and gene transcription. Curr Opin Drug Discov Devel. 2009;12(5):659-65.

129. Schmitges FW, Prusty AB, Faty M, Stutzer A, Lingaraju GM, Aiwazian J, et al.

Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell.

2011;42(3):330-41.

130. Fischle W, Wang Y, Jacobs SA, Kim Y, Allis CD, Khorasanizadeh S. Molecular

basis for the discrimination of repressive methyl-lysine marks in histone H3 by

Polycomb and HP1 chromodomains. Genes Dev. 2003;17(15):1870-81.

131. Lu TX, Hartner J, Lim EJ, Fabry V, Mingler MK, Cole ET, et al. MicroRNA-21

limits in vivo immune response-mediated activation of the IL-12/IFN-gamma

pathway, Th1 polarization, and the severity of delayed-type hypersensitivity. J

Immunol. 2011;187(6):3362-73.

132. Fitzgerald KA, Caffrey DR. Long noncoding RNAs in innate and adaptive

immunity. Curr Opin Immunol. 2014;26C:140-6.

133. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation.

Nat Rev Genet. 2004;5(7):522-31.

134. Lucas K, Raikhel AS. Insect microRNAs: biogenesis, expression profiling and

biological functions. Insect biochemistry and molecular biology. 2013;43(1):24-

38.

135. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by

adenosines, indicates that thousands of human genes are microRNA targets. Cell.

2005;120(1):15-20.

116

136. Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat

Rev Cancer. 2006;6(4):259-69.

137. Wang JW, Li K, Hellermann G, Lockey RF, Mohapatra S. Regulating the

Regulators: microRNA and Asthma. World Allergy Organ J. 2011;4(6):94-103.

138. Baumjohann D, Ansel KM. MicroRNA-mediated regulation of T helper cell

differentiation and plasticity. Nat Rev Immunol. 2013;13(9):666-78.

139. Erdel F, Rippe K. Chromatin remodelling in mammalian cells by ISWI-type

complexes--where, when and why? FEBS J. 2011;278(19):3608-18.

140. Bergman Y, Cedar H. DNA methylation dynamics in health and disease. Nat

Struct Mol Biol. 2013;20(3):274-81.

141. Arney KL, Bao S, Bannister AJ, Kouzarides T, Surani MA. Histone methylation

defines epigenetic asymmetry in the mouse zygote. Int J Dev Biol.

2002;46(3):317-20.

142. Mayer W, Niveleau A, Walter J, Fundele R, Haaf T. Demethylation of the zygotic

paternal genome. Nature. 2000;403(6769):501-2.

143. Gill ME, Erkek S, Peters AH. Parental epigenetic control of embryogenesis: a

balance between inheritance and reprogramming? Curr Opin Cell Biol.

2012;24(3):387-96.

144. Tremblay KD, Duran KL, Bartolomei MS. A 5' 2-kilobase-pair region of the

imprinted mouse H19 gene exhibits exclusive paternal methylation throughout

development. Mol Cell Biol. 1997;17(8):4322-9.

145. Lane N, Dean W, Erhardt S, Hajkova P, Surani A, Walter J, et al. Resistance of

IAPs to methylation reprogramming may provide a mechanism for epigenetic

inheritance in the mouse. Genesis. 2003;35(2):88-93.

146. Quenneville S, Verde G, Corsinotti A, Kapopoulou A, Jakobsson J, Offner S, et

al. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide

to affect chromatin and DNA methylation of imprinting control regions. Mol Cell.

2011;44(3):361-72.

147. Zuo X, Sheng J, Lau HT, McDonald CM, Andrade M, Cullen DE, et al. Zinc

finger protein ZFP57 requires its co-factor to recruit DNA methyltransferases and

maintains DNA methylation imprint in embryonic stem cells via its transcriptional

repression domain. J Biol Chem. 2012;287(3):2107-18.

148. Schultz DC, Ayyanathan K, Negorev D, Maul GG, Rauscher FJ, 3rd. SETDB1: a

novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that

117

contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-

finger proteins. Genes Dev. 2002;16(8):919-32.

149. Santos F, Hendrich B, Reik W, Dean W. Dynamic reprogramming of DNA

methylation in the early mouse embryo. Dev Biol. 2002;241(1):172-82.

150. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and

Dnmt3b are essential for de novo methylation and mammalian development. Cell.

1999;99(3):247-57.

151. Keohane AM, Lavender JS, O'Neill LP, Turner BM. Histone acetylation and X

inactivation. Dev Genet. 1998;22(1):65-73.

152. Plath K, Fang J, Mlynarczyk-Evans SK, Cao R, Worringer KA, Wang H, et al.

Role of histone H3 lysine 27 methylation in X inactivation. Science.

2003;300(5616):131-5.

153. Seki Y, Hayashi K, Itoh K, Mizugaki M, Saitou M, Matsui Y. Extensive and

orderly reprogramming of genome-wide chromatin modifications associated with

specification and early development of germ cells in mice. Dev Biol.

2005;278(2):440-58.

154. Hajkova P, Erhardt S, Lane N, Haaf T, El-Maarri O, Reik W, et al. Epigenetic

reprogramming in mouse primordial germ cells. Mech Dev. 2002;117(1-2):15-23.

155. Hajkova P, Ancelin K, Waldmann T, Lacoste N, Lange UC, Cesari F, et al.

Chromatin dynamics during epigenetic reprogramming in the mouse germ line.

Nature. 2008;452(7189):877-81.

156. Davis TL, Yang GJ, McCarrey JR, Bartolomei MS. The H19 methylation imprint

is erased and re-established differentially on the parental alleles during male germ

cell development. Hum Mol Genet. 2000;9(19):2885-94.

157. Henckel A, Chebli K, Kota SK, Arnaud P, Feil R. Transcription and histone

methylation changes correlate with imprint acquisition in male germ cells. EMBO

J. 2012;31(3):606-15.

158. Lucifero D, Mann MR, Bartolomei MS, Trasler JM. Gene-specific timing and

epigenetic memory in oocyte imprinting. Hum Mol Genet. 2004;13(8):839-49.

159. Hiura H, Obata Y, Komiyama J, Shirai M, Kono T. Oocyte growth-dependent

progression of maternal imprinting in mice. Genes Cells. 2006;11(4):353-61.

160. Liu B, Yee KM, Tahk S, Mackie R, Hsu C, Shuai K. PIAS1 SUMO ligase

regulates the self-renewal and differentiation of hematopoietic stem cells. EMBO

J. 2014;33(2):101-13.

118

161. Broske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, et al.

DNA methylation protects hematopoietic stem cell multipotency from

myeloerythroid restriction. Nat Genet. 2009;41(11):1207-15.

162. Konuma T, Oguro H, Iwama A. Role of the polycomb group proteins in

hematopoietic stem cells. Dev Growth Differ. 2010;52(6):505-16.

163. Santourlidis S, Graffmann N, Christ J, Uhrberg M. Lineage-specific transition of

histone signatures in the killer cell Ig-like receptor locus from hematopoietic

progenitor to NK cells. J Immunol. 2008;180(1):418-25.

164. Maier H, Ostraat R, Gao H, Fields S, Shinton SA, Medina KL, et al. Early B cell

factor cooperates with Runx1 and mediates epigenetic changes associated with

mb-1 transcription. Nat Immunol. 2004;5(10):1069-77.

165. Cao Y, Li H, Sun Y, Chen X, Liu H, Gao X, et al. Interferon regulatory factor 4

regulates thymocyte differentiation by repressing Runx3 expression. Eur J

Immunol. 2010;40(11):3198-209.

166. Rui J, Liu H, Zhu X, Cui Y, Liu X. Epigenetic silencing of CD8 genes by

ThPOK-mediated deacetylation during CD4 T cell differentiation. J Immunol.

2012;189(3):1380-90.

167. Kim SY, Romero R, Tarca AL, Bhatti G, Kim CJ, Lee J, et al. Methylome of fetal

and maternal monocytes and macrophages at the feto-maternal interface. Am J

Reprod Immunol. 2012;68(1):8-27.

168. White GP, Hollams EM, Yerkovich ST, Bosco A, Holt BJ, Bassami MR, et al.

CpG methylation patterns in the IFNgamma promoter in naive T cells: variations

during Th1 and Th2 differentiation and between atopics and non-atopics. Pediatr

Allergy Immunol. 2006;17(8):557-64.

169. Kaminuma O, Kitamura F, Miyatake S, Yamaoka K, Miyoshi H, Inokuma S, et al.

T-box 21 transcription factor is responsible for distorted T(H)2 differentiation in

human peripheral CD4+ T cells. J Allergy Clin Immunol. 2009;123(4):813-23 e3.

170. Weitzel RP, Lesniewski ML, Haviernik P, Kadereit S, Leahy P, Greco NJ, et al.

microRNA 184 regulates expression of NFAT1 in umbilical cord blood CD4+ T

cells. Blood. 2009;113(26):6648-57.

171. Goriely S, Van Lint C, Dadkhah R, Libin M, De Wit D, Demonte D, et al. A

defect in nucleosome remodeling prevents IL-12(p35) gene transcription in

neonatal dendritic cells. J Exp Med. 2004;199(7):1011-6.

119

172. Porras A, Kozar S, Russanova V, Salpea P, Hirai T, Sammons N, et al.

Developmental and epigenetic regulation of the human TLR3 gene. Mol

Immunol. 2008;46(1):27-36.

173. Chassin C, Kocur M, Pott J, Duerr CU, Gutle D, Lotz M, et al. miR-146a

mediates protective innate immune tolerance in the neonate intestine. Cell Host

Microbe. 2010;8(4):358-68.

174. Martino DJ, Tulic MK, Gordon L, Hodder M, Richman T, Metcalfe J, et al.

Evidence for age-related and individual-specific changes in DNA methylation

profile of mononuclear cells during early immune development in humans.

Epigenetics. 2011;6(9).

175. Nestor CE, Barrenas F, Wang H, Lentini A, Zhang H, Bruhn S, et al. DNA

methylation changes separate allergic patients from healthy controls and may

reflect altered CD4+ T-cell population structure. PLoS Genet.

2014;10(1):e1004059.

176. Morales E, Bustamante M, Vilahur N, Escaramis G, Montfort M, de Cid R, et al.

DNA hypomethylation at ALOX12 is associated with persistent wheezing in

childhood. Am J Respir Crit Care Med. 2012;185(9):937-43.

177. Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ, et al. Expression and

activity of histone deacetylases in human asthmatic airways. Am J Respir Crit

Care Med. 2002;166(3):392-6.

178. Gaffin JM, Raby BA, Petty CR, Hoffman EB, Baccarelli AA, Gold DR, et al.

beta-2 adrenergic receptor gene methylation is associated with decreased asthma

severity in inner-city schoolchildren: asthma and rhinitis. Clin Exp Allergy.

2014;44(5):681-9.

179. Liang Y, Wang P, Zhao M, Liang G, Yin H, Zhang G, et al. Demethylation of the

FCER1G promoter leads to FcepsilonRI overexpression on monocytes of patients

with atopic dermatitis. Allergy. 2012;67(3):424-30.

180. Luo Y, Zhou B, Zhao M, Tang J, Lu Q. Promoter demethylation contributes to

TSLP overexpression in skin lesions of patients with atopic dermatitis. Clin Exp

Dermatol. 2014;39(1):48-53.

181. Sonkoly E, Janson P, Majuri ML, Savinko T, Fyhrquist N, Eidsmo L, et al. MiR-

155 is overexpressed in patients with atopic dermatitis and modulates T-cell

proliferative responses by targeting cytotoxic T lymphocyte-associated antigen 4.

J Allergy Clin Immunol. 2010;126(3):581-9 e1-20.

120

182. Seumois G, Chavez L, Gerasimova A, Lienhard M, Omran N, Kalinke L, et al.

Epigenomic analysis of primary human T cells reveals enhancers associated with

TH2 memory cell differentiation and asthma susceptibility. Nat Immunol.

2014;15(8):777-88.

183. Breton CV, Byun HM, Wang X, Salam MT, Siegmund K, Gilliland FD. DNA

methylation in the arginase-nitric oxide synthase pathway is associated with

exhaled nitric oxide in children with asthma. Am J Respir Crit Care Med.

2011;184(2):191-7.

184. Runyon RS, Cachola LM, Rajeshuni N, Hunter T, Garcia M, Ahn R, et al. Asthma

discordance in twins is linked to epigenetic modifications of T cells. PLoS One.

2012;7(11):e48796.

185. Stefanowicz D, Hackett TL, Garmaroudi FS, Gunther OP, Neumann S, Sutanto

EN, et al. DNA methylation profiles of airway epithelial cells and PBMCs from

healthy, atopic and asthmatic children. PLoS One. 2012;7(9):e44213.

186. Soto-Ramirez N, Arshad SH, Holloway JW, Zhang H, Schauberger E, Ewart S, et

al. The interaction of genetic variants and DNA methylation of the interleukin-4

receptor gene increase the risk of asthma at age 18 years. Clin Epigenetics.

2013;5(1):1.

187. Perera F, Tang WY, Herbstman J, Tang D, Levin L, Miller R, et al. Relation of

DNA methylation of 5'-CpG island of ACSL3 to transplacental exposure to

airborne polycyclic aromatic hydrocarbons and childhood asthma. PLoS One.

2009;4(2):e4488.

188. Fu A, Leaderer BP, Gent JF, Leaderer D, Zhu Y. An environmental epigenetic

study of ADRB2 5'-UTR methylation and childhood asthma severity. Clin Exp

Allergy. 2012;42(11):1575-81.

189. Pascual M, Suzuki M, Isidoro-Garcia M, Padron J, Turner T, Lorente F, et al.

Epigenetic changes in B lymphocytes associated with house dust mite allergic

asthma. Epigenetics. 2011;6(9):1131-7.

190. Zhao M, Liu S, Luo S, Wu H, Tang M, Cheng W, et al. DNA methylation and

mRNA and microRNA expression of SLE CD4+ T cells correlate with disease

phenotype. J Autoimmun. 2014.

191. Koh BH, Hwang SS, Kim JY, Lee W, Kang MJ, Lee CG, et al. Th2 LCR is

essential for regulation of Th2 cytokine genes and for pathogenesis of allergic

asthma. Proc Natl Acad Sci U S A. 2010;107(23):10614-9.

121

192. Brand S, Kesper DA, Teich R, Kilic-Niebergall E, Pinkenburg O, Bothur E, et al.

DNA methylation of TH1/TH2 cytokine genes affects sensitization and progress

of experimental asthma. J Allergy Clin Immunol. 2012;129(6):1602-10 e6.

193. Allan RS, Zueva E, Cammas F, Schreiber HA, Masson V, Belz GT, et al. An

epigenetic silencing pathway controlling T helper 2 cell lineage commitment.

Nature. 2012;487(7406):249-53.

194. Kumar M, Ahmad T, Sharma A, Mabalirajan U, Kulshreshtha A, Agrawal A, et

al. Let-7 microRNA-mediated regulation of IL-13 and allergic airway

inflammation. J Allergy Clin Immunol. 2011;128(5):1077-85 e1-10.

195. Polikepahad S, Knight JM, Naghavi AO, Oplt T, Creighton CJ, Shaw C, et al.

Proinflammatory role for let-7 microRNAS in experimental asthma. J Biol Chem.

2010;285(39):30139-49.

196. Collison A, Mattes J, Plank M, Foster PS. Inhibition of house dust mite-induced

allergic airways disease by antagonism of microRNA-145 is comparable to

glucocorticoid treatment. J Allergy Clin Immunol. 2011;128(1):160-7 e4.

197. Barker DJ. The intrauterine environment and adult cardiovascular disease. Ciba

Found Symp. 1991;156:3-10; discussion -6.

198. Martino D, Prescott S. Epigenetics and prenatal influences on asthma and allergic

airways disease. Chest. 2011;139(3):640-7.

199. Fedulov AV, Kobzik L. Allergy risk is mediated by dendritic cells with congenital

epigenetic changes. Am J Respir Cell Mol Biol. 2011;44(3):285-92.

200. Martino D, Joo JE, Sexton-Oates A, Dang T, Allen K, Saffery R, et al.

Epigenome-wide association study reveals longitudinally stable DNA methylation

differences in CD4+ T cells from children with IgE-mediated food allergy.

Epigenetics. 2014;9(7):998-1006.

201. Harding JE, Johnston BM. Nutrition and fetal growth. Reprod Fertil Dev.

1995;7(3):539-47.

202. Stein AD, Zybert PA, van de Bor M, Lumey LH. Intrauterine famine exposure

and body proportions at birth: the Dutch Hunger Winter. Int J Epidemiol.

2004;33(4):831-6.

203. Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine exposure

in utero and early infancy. N Engl J Med. 1976;295(7):349-53.

204. Smith CA. Effects of maternal under nutrition upon the newborn infant in Holland

(1944-1945). J Pediatr. 1947;30(3):229-43.

122

205. Susser E, Neugebauer R, Hoek HW, Brown AS, Lin S, Labovitz D, et al.

Schizophrenia after prenatal famine. Further evidence. Archives of general

psychiatry. 1996;53(1):25-31.

206. Caron E, Ciofi P, Prevot V, Bouret SG. Alteration in neonatal nutrition causes

perturbations in hypothalamic neural circuits controlling reproductive function. J

Neurosci. 2012;32(33):11486-94.

207. Nian H, Delage B, Ho E, Dashwood RH. Modulation of histone deacetylase

activity by dietary isothiocyanates and allyl sulfides: studies with sulforaphane

and garlic organosulfur compounds. Environ Mol Mutagen. 2009;50(3):213-21.

208. Mathers JC. Session 2: Personalised nutrition. Epigenomics: a basis for

understanding individual differences? Proc Nutr Soc. 2008;67(4):390-4.

209. Li G, Kohorst JJ, Zhang W, Laritsky E, Kunde-Ramamoorthy G, Baker MS, et al.

Early postnatal nutrition determines adult physical activity and energy

expenditure in female mice. Diabetes. 2013;62(8):2773-83.

210. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, et al.

Persistent epigenetic differences associated with prenatal exposure to famine in

humans. Proc Natl Acad Sci U S A. 2008;105(44):17046-9.

211. McKay JA, Mathers JC. Diet induced epigenetic changes and their implications

for health. Acta Physiol (Oxf). 2011;202(2):103-18.

212. Choi SW, Friso S. Epigenetics: A New Bridge between Nutrition and Health. Adv

Nutr. 2010;1(1):8-16.

213. Shenderov BA. Gut indigenous microbiota and epigenetics. Microbial ecology in

health and disease. 2012;23.

214. Xue X, Feng T, Yao S, Wolf KJ, Liu CG, Liu X, et al. Microbiota downregulates

dendritic cell expression of miR-10a, which targets IL-12/IL-23p40. J Immunol.

2011;187(11):5879-86.

215. Kim GW, Gocevski G, Wu CJ, Yang XJ. Dietary, metabolic, and potentially

environmental modulation of the lysine acetylation machinery. Int J Cell Biol.

2010;2010:632739.

216. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al.

Metabolites produced by commensal bacteria promote peripheral regulatory T-cell

generation. Nature. 2013;504(7480):451-5.

217. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al.

Commensal microbe-derived butyrate induces the differentiation of colonic

regulatory T cells. Nature. 2013;504(7480):446-50.

123

218. Gueant JL, Namour F, Gueant-Rodriguez RM, Daval JL. Folate and fetal

programming: a play in epigenomics? Trends Endocrinol Metab. 2013;24(6):279-

89.

219. Christensen KE, MacKenzie RE. Mitochondrial one-carbon metabolism is

adapted to the specific needs of yeast, plants and mammals. Bioessays.

2006;28(6):595-605.

220. Weingartner J, Lotz K, Fanghanel J, Gedrange T, Bienengraber V, Proff P.

Induction and prevention of cleft lip, alveolus and palate and neural tube defects

with special consideration of B vitamins and the methylation cycle. J Orofac

Orthop. 2007;68(4):266-77.

221. Gueant JL, Gueant-Rodriguez RM, Anello G, Bosco P, Brunaud L, Romano C, et

al. Genetic determinants of folate and vitamin B12 metabolism: a common

pathway in neural tube defect and Down syndrome? Clin Chem Lab Med.

2003;41(11):1473-7.

222. Blom HJ, Smulders Y. Overview of homocysteine and folate metabolism. With

special references to cardiovascular disease and neural tube defects. J Inherit

Metab Dis. 2011;34(1):75-81.

223. Berry RJ, Li Z, Erickson JD, Li S, Moore CA, Wang H, et al. Prevention of

neural-tube defects with folic acid in China. China-U.S. Collaborative Project for

Neural Tube Defect Prevention. N Engl J Med. 1999;341(20):1485-90.

224. Czeizel AE, Dudas I. Prevention of the first occurrence of neural-tube defects by

periconceptional vitamin supplementation. N Engl J Med. 1992;327(26):1832-5.

225. Crane NT, Wilson DB, Cook DA, Lewis CJ, Yetley EA, Rader JI. Evaluating

food fortification options: general principles revisited with folic acid. Am J Public

Health. 1995;85(5):660-6.

226. Lewis CJ, Crane NT, Wilson DB, Yetley EA. Estimated folate intakes: data

updated to reflect food fortification, increased bioavailability, and dietary

supplement use. Am J Clin Nutr. 1999;70(2):198-207.

227. Quinlivan EP, Gregory JF, 3rd. Effect of food fortification on folic acid intake in

the United States. Am J Clin Nutr. 2003;77(1):221-5.

228. Dunstan JA, West C, McCarthy S, Metcalfe J, Meldrum S, Oddy WH, et al. The

relationship between maternal folate status in pregnancy, cord blood folate levels,

and allergic outcomes in early childhood. Allergy. 2012;67(1):50-7.

229. Fekete K, Berti C, Trovato M, Lohner S, Dullemeijer C, Souverein OW, et al.

Effect of folate intake on health outcomes in pregnancy: a systematic review and

124

meta-analysis on birth weight, placental weight and length of gestation. Nutr J.

2012;11:75.

230. Fainaru O, Shseyov D, Hantisteanu S, Groner Y. Accelerated chemokine receptor

7-mediated dendritic cell migration in Runx3 knockout mice and the spontaneous

development of asthma-like disease. Proc Natl Acad Sci U S A.

2005;102(30):10598-603.

231. Morgan HD, Sutherland HG, Martin DI, Whitelaw E. Epigenetic inheritance at

the agouti locus in the mouse. Nat Genet. 1999;23(3):314-8.

232. Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl

supplements affect agouti gene expression in Avy/a mice. FASEB J.

1998;12(11):949-57.

233. Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional

effects on epigenetic gene regulation. Mol Cell Biol. 2003;23(15):5293-300.

234. Kiefte-de Jong JC, Timmermans S, Jaddoe VW, Hofman A, Tiemeier H, Steegers

EA, et al. High circulating folate and vitamin B-12 concentrations in women

during pregnancy are associated with increased prevalence of atopic dermatitis in

their offspring. J Nutr. 2012;142(4):731-8.

235. Whitrow MJ, Moore VM, Rumbold AR, Davies MJ. Effect of supplemental folic

acid in pregnancy on childhood asthma: a prospective birth cohort study. Am J

Epidemiol. 2009;170(12):1486-93.

236. Bekkers MB, Elstgeest LE, Scholtens S, Haveman-Nies A, de Jongste JC,

Kerkhof M, et al. Maternal use of folic acid supplements during pregnancy, and

childhood respiratory health and atopy. Eur Respir J. 2012;39(6):1468-74.

237. Magdelijns FJ, Mommers M, Penders J, Smits L, Thijs C. Folic acid use in

pregnancy and the development of atopy, asthma, and lung function in childhood.

Pediatrics. 2011;128(1):e135-44.

238. Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, Lindemans J, Siebel C,

Steegers EA, et al. Periconceptional maternal folic acid use of 400 microg per day

is related to increased methylation of the IGF2 gene in the very young child. PLoS

One. 2009;4(11):e7845.

239. Hoyo C, Murtha AP, Schildkraut JM, Jirtle RL, Demark-Wahnefried W, Forman

MR, et al. Methylation variation at IGF2 differentially methylated regions and

maternal folic acid use before and during pregnancy. Epigenetics. 2011;6(7):928-

36.

125

240. McKay JA, Groom A, Potter C, Coneyworth LJ, Ford D, Mathers JC, et al.

Genetic and non-genetic influences during pregnancy on infant global and site

specific DNA methylation: role for folate gene variants and vitamin B12. PLoS

One. 2012;7(3):e33290.

241. Ba Y, Yu H, Liu F, Geng X, Zhu C, Zhu Q, et al. Relationship of folate, vitamin

B12 and methylation of insulin-like growth factor-II in maternal and cord blood.

Eur J Clin Nutr. 2011;65(4):480-5.

242. Fryer AA, Nafee TM, Ismail KM, Carroll WD, Emes RD, Farrell WE. LINE-1

DNA methylation is inversely correlated with cord plasma homocysteine in man:

a preliminary study. Epigenetics. 2009;4(6):394-8.

243. Boeke CE, Baccarelli A, Kleinman KP, Burris HH, Litonjua AA, Rifas-Shiman

SL, et al. Gestational intake of methyl donors and global LINE-1 DNA

methylation in maternal and cord blood: prospective results from a folate-replete

population. Epigenetics. 2012;7(3):253-60.

244. Fryer AA, Emes RD, Ismail KM, Haworth KE, Mein C, Carroll WD, et al.

Quantitative, high-resolution epigenetic profiling of CpG loci identifies

associations with cord blood plasma homocysteine and birth weight in humans.

Epigenetics. 2011;6(1):86-94.

245. Chang H, Zhang T, Zhang Z, Bao R, Fu C, Wang Z, et al. Tissue-specific

distribution of aberrant DNA methylation associated with maternal low-folate

status in human neural tube defects. J Nutr Biochem. 2011;22(12):1172-7.

246. Simopoulos AP. The importance of the ratio of omega-6/omega-3 essential fatty

acids. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

2002;56(8):365-79.

247. Prescott SL, Irvine J, Dunstan JA, Hii C, Ferrante A. Protein kinase Czeta: a novel

protective neonatal T-cell marker that can be upregulated by allergy prevention

strategies. J Allergy Clin Immunol. 2007;120(1):200-6.

248. Noakes PS, Vlachava M, Kremmyda LS, Diaper ND, Miles EA, Erlewyn-

Lajeunesse M, et al. Increased intake of oily fish in pregnancy: effects on neonatal

immune responses and on clinical outcomes in infants at 6 mo. Am J Clin Nutr.

2012;95(2):395-404.

249. Childs CE, Romijn T, Enke U, Hoile S, Calder PC. Maternal diet during

pregnancy has tissue-specific effects upon fetal fatty acid composition and alters

fetal immune parameters. Prostaglandins Leukot Essent Fatty Acids. 2010;83(4-

6):179-84.

126

250. Calder PC. Mechanisms of action of (n-3) fatty acids. J Nutr. 2012;142(3):592S-

9S.

251. Hoile SP, Irvine NA, Kelsall CJ, Sibbons C, Feunteun A, Collister A, et al.

Maternal fat intake in rats alters 20:4n-6 and 22:6n-3 status and the epigenetic

regulation of Fads2 in offspring liver. J Nutr Biochem. 2013;24(7):1213-20.

252. Niculescu MD, Lupu DS, Craciunescu CN. Perinatal manipulation of alpha-

linolenic acid intake induces epigenetic changes in maternal and offspring livers.

FASEB J. 2013;27(1):350-8.

253. Lee HS, Barraza-Villarreal A, Hernandez-Vargas H, Sly PD, Biessy C,

Ramakrishnan U, et al. Modulation of DNA methylation states and infant immune

system by dietary supplementation with omega-3 PUFA during pregnancy in an

intervention study. Am J Clin Nutr. 2013;98(2):480-7.

254. Magnusson LL, Olesen AB, Wennborg H, Olsen J. Wheezing, asthma, hayfever,

and atopic eczema in childhood following exposure to tobacco smoke in fetal life.

Clin Exp Allergy. 2005;35(12):1550-6.

255. Bisgaard H, Loland L, Holst KK, Pipper CB. Prenatal determinants of neonatal

lung function in high-risk newborns. J Allergy Clin Immunol. 2009;123(3):651-7,

7 e1-4.

256. Sears MR, Holdaway MD, Flannery EM, Herbison GP, Silva PA. Parental and

neonatal risk factors for atopy, airway hyper-responsiveness, and asthma. Arch

Dis Child. 1996;75(5):392-8.

257. Li YF, Langholz B, Salam MT, Gilliland FD. Maternal and grandmaternal

smoking patterns are associated with early childhood asthma. Chest.

2005;127(4):1232-41.

258. Joubert BR, Haberg SE, Nilsen RM, Wang X, Vollset SE, Murphy SK, et al.

450K epigenome-wide scan identifies differential DNA methylation in newborns

related to maternal smoking during pregnancy. Environ Health Perspect.

2012;120(10):1425-31.

259. Novakovic B, Ryan J, Pereira N, Boughton B, Craig JM, Saffery R. Postnatal

stability, tissue, and time specific effects of AHRR methylation change in

response to maternal smoking in pregnancy. Epigenetics. 2014;9(3):377-86.

260. Monick MM, Beach SR, Plume J, Sears R, Gerrard M, Brody GH, et al.

Coordinated changes in AHRR methylation in lymphoblasts and pulmonary

macrophages from smokers. Am J Med Genet B Neuropsychiatr Genet.

2012;159B(2):141-51.

127

261. Shenker NS, Polidoro S, van Veldhoven K, Sacerdote C, Ricceri F, Birrell MA, et

al. Epigenome-wide association study in the European Prospective Investigation

into Cancer and Nutrition (EPIC-Turin) identifies novel genetic loci associated

with smoking. Hum Mol Genet. 2013;22(5):843-51.

262. Philibert RA, Beach SR, Brody GH. Demethylation of the aryl hydrocarbon

receptor repressor as a biomarker for nascent smokers. Epigenetics.

2012;7(11):1331-8.

263. Zeilinger S, Kuhnel B, Klopp N, Baurecht H, Kleinschmidt A, Gieger C, et al.

Tobacco smoking leads to extensive genome-wide changes in DNA methylation.

PLoS One. 2013;8(5):e63812.

264. Toledo-Rodriguez M, Lotfipour S, Leonard G, Perron M, Richer L, Veillette S, et

al. Maternal smoking during pregnancy is associated with epigenetic

modifications of the brain-derived neurotrophic factor-6 exon in adolescent

offspring. Am J Med Genet B Neuropsychiatr Genet. 2010;153B(7):1350-4.

265. Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD. Prenatal tobacco

smoke exposure affects global and gene-specific DNA methylation. Am J Respir

Crit Care Med. 2009;180(5):462-7.

266. Patil VK, Holloway JW, Zhang H, Soto-Ramirez N, Ewart S, Arshad SH, et al.

Interaction of prenatal maternal smoking, interleukin 13 genetic variants and

DNA methylation influencing airflow and airway reactivity. Clin Epigenetics.

2013;5(1):22.

267. Liu G, Niu Z, Van Niekerk D, Xue J, Zheng L. Polycyclic aromatic hydrocarbons

(PAHs) from coal combustion: emissions, analysis, and toxicology. Rev Environ

Contam Toxicol. 2008;192:1-28.

268. Simko P. Factors affecting elimination of polycyclic aromatic hydrocarbons from

smoked meat foods and liquid smoke flavorings. Mol Nutr Food Res.

2005;49(7):637-47.

269. Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, Rudel RA.

Environmental pollutants and breast cancer: epidemiologic studies. Cancer.

2007;109(12 Suppl):2667-711.

270. Factor P, Akhmedov AT, McDonald JD, Qu A, Wu J, Jiang H, et al. Polycyclic

Aromatic Hydrocarbons Impair 2AR Function in Airway Epithelial and Smooth

Muscle Cells. Am J Respir Cell Mol Biol. 2011.

128

271. Choi H, Jedrychowski W, Spengler J, Camann DE, Whyatt RM, Rauh V, et al.

International studies of prenatal exposure to polycyclic aromatic hydrocarbons

and fetal growth. Environ Health Perspect. 2006;114(11):1744-50.

272. Perera FP, Rauh V, Whyatt RM, Tang D, Tsai WY, Bernert JT, et al. A summary

of recent findings on birth outcomes and developmental effects of prenatal ETS,

PAH, and pesticide exposures. Neurotoxicology. 2005;26(4):573-87.

273. Tang WY, Levin L, Talaska G, Cheung YY, Herbstman J, Tang D, et al. Maternal

exposure to polycyclic aromatic hydrocarbons and 5'-CpG methylation of

interferon-gamma in cord white blood cells. Environ Health Perspect.

2012;120(8):1195-200.

274. Liu J, Ballaney M, Al-alem U, Quan C, Jin X, Perera F, et al. Combined inhaled

diesel exhaust particles and allergen exposure alter methylation of T helper genes

and IgE production in vivo. Toxicol Sci. 2008;102(1):76-81.

275. Gilmour PS, Rahman I, Donaldson K, MacNee W. Histone acetylation regulates

epithelial IL-8 release mediated by oxidative stress from environmental particles.

Am J Physiol Lung Cell Mol Physiol. 2003;284(3):L533-40.

276. Michel S, Busato F, Genuneit J, Pekkanen J, Dalphin JC, Riedler J, et al. Farm

exposure and time trends in early childhood may influence DNA methylation in

genes related to asthma and allergy. Allergy. 2013;68(3):355-64.

277. Slaats GG, Reinius LE, Alm J, Kere J, Scheynius A, Joerink M. DNA methylation

levels within the CD14 promoter region are lower in placentas of mothers living

on a farm. Allergy. 2012;67(7):895-903.

278. Schaub B, Liu J, Hoppler S, Schleich I, Huehn J, Olek S, et al. Maternal farm

exposure modulates neonatal immune mechanisms through regulatory T cells. J

Allergy Clin Immunol. 2009;123(4):774-82 e5.

279. Douwes J, Cheng S, Travier N, Cohet C, Niesink A, McKenzie J, et al. Farm

exposure in utero may protect against asthma, hay fever and eczema. Eur Respir J.

2008;32(3):603-11.

280. Brand S, Teich R, Dicke T, Harb H, Yildirim AO, Tost J, et al. Epigenetic

regulation in murine offspring as a novel mechanism for transmaternal asthma

protection induced by microbes. J Allergy Clin Immunol. 2011;128(3):618-25 e1-

7.

281. Houseman EA, Molitor J, Marsit CJ. Reference-free cell mixture adjustments in

analysis of DNA methylation data. Bioinformatics. 2014;30(10):1431-9.

129

282. Toperoff G, Aran D, Kark JD, Rosenberg M, Dubnikov T, Nissan B, et al.

Genome-wide survey reveals predisposing diabetes type 2-related DNA

methylation variations in human peripheral blood. Hum Mol Genet.

2012;21(2):371-83.

283. Rakyan VK, Beyan H, Down TA, Hawa MI, Maslau S, Aden D, et al.

Identification of type 1 diabetes-associated DNA methylation variable positions

that precede disease diagnosis. PLoS Genet. 2011;7(9):e1002300.

284. Waterland RA, Kellermayer R, Laritsky E, Rayco-Solon P, Harris RA, Travisano

M, et al. Season of conception in rural gambia affects DNA methylation at

putative human metastable epialleles. PLoS Genet. 2010;6(12):e1001252.

285. Meagher RB, Mussar KJ. The influence of DNA sequence on epigenome-induced

pathologies. Epigenetics & chromatin. 2012;5(1):11.

286. Laird PW. Principles and challenges of genomewide DNA methylation analysis.

Nat Rev Genet. 2010;11(3):191-203.

287. Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies

for common human diseases. Nat Rev Genet. 2011;12(8):529-41.

288. Dunstan JA, Prescott SL. Does fish oil supplementation in pregnancy reduce the

risk of allergic disease in infants? Curr Opin Allergy Clin Immunol.

2005;5(3):215-21.

289. D'Vaz N, Meldrum SJ, Dunstan JA, Lee-Pullen TF, Metcalfe J, Holt BJ, et al.

Fish oil supplementation in early infancy modulates developing infant immune

responses. Clin Exp Allergy. 2012;42(8):1206-16.

290. Pfeiffer CM, Caudill SP, Gunter EW, Osterloh J, Sampson EJ. Biochemical

indicators of B vitamin status in the US population after folic acid fortification:

results from the National Health and Nutrition Examination Survey 1999-2000.

Am J Clin Nutr. 2005;82(2):442-50.

291. Shane B. Folate fortification: enough already? Am J Clin Nutr. 2003;77(1):8-9.

292. Haberg SE, London SJ, Stigum H, Nafstad P, Nystad W. Folic acid supplements

in pregnancy and early childhood respiratory health. Arch Dis Child.

2009;94(3):180-4.

293. Sadli N, Ackland ML, De Mel D, Sinclair AJ, Suphioglu C. Effects of zinc and

DHA on the epigenetic regulation of human neuronal cells. Cell Physiol Biochem.

2012;29(1-2):87-98.

294. Ceccarelli V, Racanicchi S, Martelli MP, Nocentini G, Fettucciari K, Riccardi C,

et al. Eicosapentaenoic acid demethylates a single CpG that mediates expression

130

of tumor suppressor CCAAT/enhancer-binding protein delta in U937 leukemia

cells. J Biol Chem. 2011;286(31):27092-102.

295. Dreborg S. The skin prick test in the diagnosis of atopic allergy. Journal of the

American Academy of Dermatology. 1989;21(4 Pt 2):820-1.

296. Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of

the European Task Force on Atopic Dermatitis. Dermatology. 1993;186(1):23-31.

297. Prescott SL, Macaubas C, Holt BJ, Smallacombe TB, Loh R, Sly PD, et al.

Transplacental priming of the human immune system to environmental allergens:

universal skewing of initial T cell responses toward the Th2 cytokine profile. J

Immunol. 1998;160(10):4730-7.

298. Martino D, Joo JE, Sexton-Oates A, Dang T, Allen K, Saffery R, et al.

Epigenome-wide association study reveals longitudinally stable DNA methylation

differences in CD4+ T cells from children with IgE-mediated food allergy.

Epigenetics. 2014;9(7).

299. Hung CH, Yang SN, Kuo PL, Chu YT, Chang HW, Wei WJ, et al. Modulation of

cytokine expression in human myeloid dendritic cells by environmental

endocrine-disrupting chemicals involves epigenetic regulation. Environ Health

Perspect. 2010;118(1):67-72.

300. Gunawardhana LP, Gibson PG, Simpson JL, Benton MC, Lea RA, Baines KJ.

Characteristic DNA methylation profiles in peripheral blood monocytes are

associated with inflammatory phenotypes of asthma. Epigenetics. 2014;9(9):1302-

16.

301. Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, et

al. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina

Infinium HumanMethylation450 microarray. Epigenetics. 2013;8(2):203-9.

302. Butler MG. Genomic imprinting disorders in humans: a mini-review. Journal of

assisted reproduction and genetics. 2009;26(9-10):477-86.

303. Plant K, Fairfax BP, Makino S, Vandiedonck C, Radhakrishnan J, Knight JC. Fine

mapping genetic determinants of the highly variably expressed MHC gene ZFP57.

Eur J Hum Genet. 2013.

304. Tada Y, Yamaguchi Y, Kinjo T, Song X, Akagi T, Takamura H, et al. The stem

cell transcription factor ZFP57 induces IGF2 expression to promote anchorage-

independent growth in cancer cells. Oncogene. 2014.

131

305. Ladd-Acosta C, Hansen KD, Briem E, Fallin MD, Kaufmann WE, Feinberg AP.

Common DNA methylation alterations in multiple brain regions in autism.

Molecular psychiatry. 2014;19(8):862-71.

306. Wilson CB, Rowell E, Sekimata M. Epigenetic control of T-helper-cell

differentiation. Nat Rev Immunol. 2009;9(2):91-105.

307. Pan HF, Leng RX, Li XP, Zheng SG, Ye DQ. Targeting T-helper 9 cells and

interleukin-9 in autoimmune diseases. Cytokine & growth factor reviews.

2013;24(6):515-22.

308. Brough HA, Cousins DJ, Munteanu A, Wong YF, Sudra A, Makinson K, et al. IL-

9 is a key component of memory T cell peanut-specific responses from children

with peanut allergy. J Allergy Clin Immunol. 2014.

309. Gabory A, Ferry L, Fajardy I, Jouneau L, Gothie JD, Vige A, et al. Maternal diets

trigger sex-specific divergent trajectories of gene expression and epigenetic

systems in mouse placenta. PLoS One. 2012;7(11):e47986.

310. Masuyama H, Hiramatsu Y. Effects of a high-fat diet exposure in utero on the

metabolic syndrome-like phenomenon in mouse offspring through epigenetic

changes in adipocytokine gene expression. Endocrinology. 2012;153(6):2823-30.

311. Kryukov GV, Shpunt A, Stamatoyannopoulos JA, Sunyaev SR. Power of deep,

all-exon resequencing for discovery of human trait genes. Proc Natl Acad Sci U S

A. 2009;106(10):3871-6.

312. Lander ES, Botstein D. Mapping mendelian factors underlying quantitative traits

using RFLP linkage maps. Genetics. 1989;121(1):185-99.

313. Van Gestel S, Houwing-Duistermaat JJ, Adolfsson R, van Duijn CM, Van

Broeckhoven C. Power of selective genotyping in genetic association analyses of

quantitative traits. Behavior genetics. 2000;30(2):141-6.

314. Guey LT, Kravic J, Melander O, Burtt NP, Laramie JM, Lyssenko V, et al. Power

in the phenotypic extremes: a simulation study of power in discovery and

replication of rare variants. Genet Epidemiol. 2011.

315. Myles IA, Pincus NB, Fontecilla NM, Datta SK. Effects of parental omega-3 fatty

acid intake on offspring microbiome and immunity. PLoS One. 2014;9(1):e87181.

316. Myles IA, Fontecilla NM, Janelsins BM, Vithayathil PJ, Segre JA, Datta SK.

Parental dietary fat intake alters offspring microbiome and immunity. J Immunol.

2013;191(6):3200-9.

317. Hallam MC, Barile D, Meyrand M, German JB, Reimer RA. Maternal high-

protein or high-prebiotic-fiber diets affect maternal milk composition and gut

132

microbiota in rat dams and their offspring. Obesity (Silver Spring).

2014;22(11):2344-51.

318. Ma J, Prince AL, Bader D, Hu M, Ganu R, Baquero K, et al. High-fat maternal

diet during pregnancy persistently alters the offspring microbiome in a primate

model. Nat Commun. 2014;5:3889.

319. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta

harbors a unique microbiome. Sci Transl Med. 2014;6(237):237ra65.

133

APPENDIX 1

BUFFER RECIPIES

PBS – Citrate (0.6%)

500ml 1x PBS

3g tri-sodium citrate dihydrate

PCF

50ml PBS-Citrate

1ml FCS (heat inactivated)

PCFD

5ml PCF

50ul DNAse (1mg/ml)

PBS-BSA (1%)

100ml PBS

1g BSA

FACS buffer

500ml PBS

2.5g BSA

134

APPENDIX 2

Primer sequences used in validation studies in Chapter 3

Validation of candidate loci by Sequenome EpiTYPER platform Primer name Tag + Sequence Chromosome 6 DMR – Left primer

aggaagagagGAGGATTTTAGAGGTTGGAAGTTTT

Chromosome 6 DMR – Right primer

cagtaatacgactcactatagggagaaggctCCCTCTCATCTAAATCAAAAAACAC

Primers for H3/H4 acetylation by Chromatin Immunoprecipitation (ChIP) Primers for ZFP57 transcripts (Ensemble)

Sequence Sequence start and end sites relative to the transcription start site

ZFP57-001 distal (ZFP57_1D) - forward

cac cac cgg cta act ttt gt starts at bp -801 and ends at bp -545

- reverse cct ggg caa aaa gag tga aa

ZFP57-002 proximal (ZFP57_2P) - forward

ccc agg ctg gtg ttg tta ct starts at bp -173 and ends at bp +20

-reverse

ggt ttg atg tgg ctt cct gt

ZFP57-002 distal (ZFP57_2D) - forward

cat gga aga gat ctt aga gag tg starts at bp -603 and ends at -366

reverse acc taa tgc aga gat gct aaa taa

135

Human promoter primers used for the acetylation experiments in Chapter 5

Locus Forward Reverse

RPL32 5‘-GGAAGTGCTTGCCTTTTTCC-3‘ 5‘-GGATTGCCACGGATTAACAC-3‘

IL1B 5‘-CGTGGGAAAATCCAGTATTTTAATG-3‘

5‘-CAAATGTATCACCATGCAAATATGC-3‘

IL4 5‘-TGGGTAAGGACCTTATGGACC-3‘ 5‘-GGTGGCATCTTGGAAACTGTC-3‘

IL5 5‘-AGGAGATCTTTTTAGTCACTGGCAACA-3‘

5‘-CGTCTCGAGGGCAAAGAAAGTGCATAG-3‘

IL9 5‘-CGTTAGAACACCCATGAC-3‘ 5‘-TTCTGGTTGTGAGAGTTAG-3‘

IL10 5‘-GACAACACTACTAAGGCTCCTTTGGGA-3‘

5‘-GTGAGCAAACTGAGGCACAGAAAT-3‘

IL13 5‘-TGTGGGAGATGCCGTGGG-3‘ 5‘-TCTGACTCCCAGAAGTCTGC-3‘

IL17A 5‘-AATTTCTGCCCTTCCCATTT-3‘ 5‘-CCCAGGAGTCATCGTTGTTT-3‘

GATA3 5‘-CACATTTAAAGGGCCAGAGC-3‘ 5‘-AAGGAAACTGCAACCCAAAC-3‘

TBX21 5‘-TGGCCAAGAGCGTAGAATTT-3‘ 5‘-GCTTTGCTGTGGCTTTATGA-3‘

RORC 5‘-TCTCCCCTATGCCTGTCACCTG-3‘ 5‘-TGATTTTGCCCAAGGACTCACAC-3‘

FOP3 5‘-ATCGTGAGGATGGATGCATTAATA-3‘ 5‘-CCACTGGGAAGGTCCCTAGC-3‘

IRF1 5‘-GTACTTCCCCTTCGCCG-3‘ 5‘-GCGTACTCACCTCTGCTGC-3‘

TNF 5‘-AGAAGACCCCCCTCGGAACC-3‘ 5‘-ATCTGGAGGAAGCGGTAGTG-3‘

IFNG 5‘-AATCCCACCAGAATGGCACAGGTG-3‘

5‘-GAACAATGTGCTGCACCTCCTCTGG-3‘

MS4A2 5‘-CTTAGGGGTTAGATTTTATGTGTTTGAACCCCAA-3‘

5‘-CCATCTTCTTCATGGACTCCTGGTGCTTAC-3‘

136

The reference ranges for lower limit of detection (LOD), sensitivity and specificity

for analysed genes for ChIP in Chapter 5

137

APPENDIX 3

Purity of isolated cells by magnetic bead separation

1st

experiment 2nd

experiment 3rd

experiment 4th

experiment PBMC before subjected to magnetic bead separation CD4 44.3 26.6 50.7 48.9 CD8 20.7 24.8 11.8 11.4 CD14 11.5 18.4 18.9 14.5 CD19 4.54 6.11 6.7 6.4 Extracted CD4 CD4 95.9 92.3 90.5 89.6 CD8 0.054 0 0.03 0.0 CD14 0.481 4.51 6.41 4.9 CD19 3.08 1.11 2.43 0.6 Extracted CD8 CD4 1.47 4.31 10.4 17.9 CD8 92.8 79.5 78.8 62.7 CD14 0.029 2.6 2.38 3.4 CD19 3.63 3.95 2.43 2.5

Remaining cell mix after magnetic bead separation CD3 7.62 17.1 3.6 CD14 25.7 35.7 51.1 CD19 4.76 8.42 11.3