maternal high fat diet alters the transcriptional … · board games, brunch and dinner nights,...

117
Maternal High Fat Diet Alters the Transcriptional Response to Glucocorticoid and Immune Challenge in the Amygdala and Hippocampus of Adult Rat Offspring by Christine Min Wei Lum A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Cell & Systems Biology University of Toronto © Copyright by Christine Min Wei Lum 2016

Upload: lediep

Post on 13-Aug-2018

214 views

Category:

Documents


0 download

TRANSCRIPT

Maternal High Fat Diet Alters the Transcriptional Response to

Glucocorticoid and Immune Challenge in the Amygdala and

Hippocampus of Adult Rat Offspring

by

Christine Min Wei Lum

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Graduate Department of Cell & Systems Biology

University of Toronto

© Copyright by Christine Min Wei Lum 2016

ii

Maternal High Fat Diet Alters the Transcriptional Response to Glucocorticoid and Immune

Challenge in the Amygdala and Hippocampus of Adult Rat Offspring

Master of Science 2016, Christine Min Wei Lum, Cell & Systems Biology, University of

Toronto

Abstract

Maternal overnutrition during pre- and post-(peri)natal periods of development has previously

been suggested to increase offspring risk for developing psychiatric disorders. The mental

impacts are often accompanied by altered expressions of glucocorticoid receptors (GR), which

regulate the inflammatory response to stress. In this study, rats were acutely injected with

corticosterone (CORT) and/or lipopolysaccharide (LPS) in adulthood after perinatal exposure to

maternal high-fat diet (HFD) to examine its effects on responding to stress. Gene transcript

analyses in the amygdala and hippocampus revealed higher immunosuppressive activity and

altered abundance of genes associated with anxiety- and depressive-like behavior compared to

rats perinatally exposed to standard diets. These effects were sex and brain region-specific, and

appeared to coincide with levels of GR. Epigenetic analyses suggest impairments in the DNA

methylation machinery to be related to these diet differences. Hence, perinatal HFD incurs

differential immune responses to stress in the rodent brain.

iii

Acknowledgements

I would like to thank all my friends back home, and at UTSC for offering me their kind, listening

ear, emotional support, comedic relief, and place of refuge. Thank you all for reminding me that

even though no one's perfect, and that everyone has needs which may conflict with our own,

most people want to help and empathize, and that every small gesture counts.

I would also like to thank my family for providing me a steady supply of energy to keep me

going through each and every day, and for always trying to adapt to my needs. I want to give a

special shout-out to my sister Jocelyn for showing me that I will always be loved no matter

where I am, and who I become as I keep trying to move forward.

Finally, I want to thank my lab mates for the good times we've had together ex. the road trips,

board games, brunch and dinner nights, Lego movie - and for the rare times in my life where I

have truly, deeply been candid with another person, and have felt that I was known and cherished

in return...whatever comes next.

iv

Contributions

I am grateful to the students and faculty at UTSC for their help with the following:

Professor Patrick McGowan - Supervision, mentorship, provision of great ideas and direction,

sacrifice of rats and brain extractions.

Wilfred de Vega, PhD Candidate - Sacrifice of rats and brain extractions, brain microdissections,

portion of qPCRs for LPS condition, evaluation of housekeeping gene stability with geNorm (R-

package), general assistance.

Sameera Abuaish, MSc - Brain microdissections.

Shathvee Sivanathan, Atif Hussain, Patrick Ng, BSc - Handling and weighing of rats, maternal

behavioral coding, drug injections.

Ben Hing, PhD; Professors Mauricio Terebiznik, Joanne Nash - Excellent research advice and

provision of critical thought.

Aya Sasaki, MSc - Sacrifice of rats and brain extractions, teaching brain microdissections.

Pauline Pan and Sophie St-Cyr, MSc - Technical advice on DNA cleanup.

v

Table of Contents

Acknowledgments..........................................................................................................................iii

Contributions..................................................................................................................................iv

Table of Contents.............................................................................................................................v

List of Tables..................................................................................................................................ix

List of Figures..................................................................................................................................x

List of Supplementary Tables.........................................................................................................xi

List of Abbreviations.....................................................................................................................xii

Chapter 1 Introduction.....................................................................................................................1

1.1 Effects of Maternal Obesity and Overnutrition on Offspring physical and mental

health....................................................................................................................................2

1.2 Perinatal Programming of the HPA-axis: Effect on Mental Health..............................4

1.2.1 Effects of GR and MR on Anxiety and Depression........................................7

1.2.2 Overexposure to GCs alters GR and MR expression......................................8

1.3 Mediation of CORT and LPS-induced Inflammatory Changes Through GR and

MR..................................................... .................................................................................8

1.3.1 The General Immunosuppressive Action of GCs...........................................9

1.3.2 Inflammatory Actions of GCs in the Central Nervous System (CNS).........11

vi

1.4 Neuroinflammation, GR, and Mental Disorders: Implications of Maternal

Overnutrition......................................................................................................................13

1.4.1 How Cytokines in the CNS Promote Psychopathology................................14

1.4.2 Caveats to Consider with Maternal Overnutrition Studies...........................16

1.4.3 Effects of Perinatal HFD on Inflammation...................................................16

1.4.4 Effects of Perinatal HFD on the HPA-axis as a Consequence of

Inflammation..........................................................................................................18

1.5 Role of Epigenetic Regulation in Incurring Lifelong Effects of Maternal Over

Nutrition ............................................................................................................................19

Box 1. Regulators of DNA methylation................................................................20

1.6 Role of Epigenetic Regulation in Responding to Stress..............................................21

1.6.1 Gene-specific Functions................................................................................21

1.6.2 Mediation of Global Changes.......................................................................22

1.7 The Effects of Prior Experiences on Responses to Stress............................................22

1.7.1 Studies inspecting HPA-axis Activity..........................................................23

1.7.2 Studies examining Transcriptional and Epigenetic effects...........................24

1.7.3 Other studies with Inflammatory Pathways..................................................25

1.8 Research Hypotheses...................................................................................................25

vii

Chapter 2 Materials & Methods.....................................................................................................28

2.1 Animals........................................................................................................................28

2.2 Diets.............................................................................................................................28

2.3 Subjects and general procedures..................................................................................29

2.4 Drug challenges...........................................................................................................29

2.5 Brain tissue preparation...............................................................................................30

2.6 Analyses of transcript abundance by quantitative real-time reverse transcriptase-

polymerase chain reaction (qRT-PCR)..............................................................................31

2.7 Primer specifications....................................................................................................33

2.8 Measurement of DNMT enzyme activity and abundance in the amygdala.................34

2.9 Global CpG methylation analysis in the amygdala with Luminometric methylation

assay (LUMA) ..................................................................................................................34

2.10 Statistical analyses.....................................................................................................35

Chapter 3 Results...........................................................................................................................36

3.1 Perinatal-HFD offspring display differences in body weight throughout

development.......................................................................................................................36

3.2 Perinatal-HFD exposed animals display exacerbated anti-inflammatory transcriptional

responses to CORT challenge in the amygdala.................................................................37

viii

3.3 Perinatal-HFD exposure incurs similar, but sex-dependent transcriptional responses to

CORT in the hippocampus.................................................................................................41

3.4 Perinatal HFD-exposed animals display exacerbated anti-inflammatory transcriptional

responses to LPS challenge in the amygdala.....................................................................45

3.5 Perinatal-HFD exposed animals display altered pro-inflammatory transcriptional

responses to CORT/LPS challenge in the amygdala.........................................................48

3.6 Diet-dependent transcriptional responses cluster with transcript levels of epigenetic

regulators in the amygdala.................................................................................................52

3.7 DNMT activity reduces in response to CORT and CORT/LPS for Perinatal HFD-

exposed males, but not CORT/LPS females in the amygdala...........................................53

3.8 DNMT activity correlates with global CpG methylation in CORT-injected male

groups in the amygdala......................................................................................................56

Chapter 4 Discussion.....................................................................................................................57

4.1 Interpretation of results from CORT challenge...........................................................57

4.1.1 Diet x Sex differences in the Amygdala.......................................................57

4.1.2 Females' distinct response in the Hippocampus...........................................61

4.2 Results from LPS challenge in the amygdala..............................................................62

4.3 Results from CORT/LPS challenge in the amygdala..................................................63

4.4 Comparison of results with Sasaki et al. (2013) Maternal HFD Study.......................65

ix

4.5 Maternal HFD transmission of CORT as possible mechanism for differential GC

sensitivity...........................................................................................................................67

4.6 Role of Epigenetic Regulators: MeCP2 as potential hub for manifestation of diet- and

sex- differences..................................................................................................................68

4.7 Explaining the effects of DNMT activity on Global CpG methylation levels............70

4.8 Conclusion...................................................................................................................71

References.....................................................................................................................................71

Appendix......................................................................................................................................101

List of Tables

Table 1 Number of adult offspring in each challenge condition...................................................30

Table 2 List of genes analyzed for transcript abundance with qPCR............................................32

Table 3 Primer sequences and catalogue numbers for genes analyzed with qPCR.......................33

Table 4 Transcriptional response to CORT challenge in the amygdala........................................40

Table 5 Transcriptional response to CORT challenge in the hippocampus...................................44

Table 6 Transcriptional response to LPS challenge in the amygdala............................................47

Table 7 Transcriptional response to CORT/LPS challenge in the amygdala................................51

x

Table 8 Transcriptional response to CORT and CORT/LPS in the amygdala grouped by post-hoc

comparisons within drug................................................................................................................52

Table 9 Spearman's Correlation Tests Between DNMT transcript abundance and Total DNMT

activity............................................................................................................................................55

List of Figures

Figure 1 Summary of the HPA-axis from Smith and Vale (2006) ...............................................6

Figure 2 Simplified diagram of the general CORT/LPS pathway.................................................10

Figure 3 Perinatal-HFD regimen used in this study.......................................................................29

Figure 4 Body weights of offspring...............................................................................................36

Figure 5 Transcript level data for CORT-challenged males in the amygdala...............................38

Figure 6 Transcript level data for CORT-challenged females in the amygdala............................39

Figure 7 Transcript level data for CORT-challenged males in the hippocampus..........................42

Figure 8 Transcript level data for CORT-challenged females in the hippocampus......................43

Figure 9 Transcript level data for LPS-challenged males in the amygdala...................................45

Figure 10 Transcript level data for LPS-challenged females in the amygdala..............................46

Figure 11 Increases in IL-6/IL-10 ratios between LPS and CORT/LPS.......................................48

Figure 12. Transcript level data for CORT/LPS-challenged males in the amygdala....................49

xi

Figure 13 Transcript level data for CORT/LPS-challenged females in the amygdala..................50

Figure 14 DNMT Activity and Protein Assays.............................................................................54

Figure 15 Global CpG methylation measured by LUMA in the amygdala of CORT-injected

males..............................................................................................................................................56

Figure 16 Comparison of IL-6/IL-10 ratios between CORT-challenged males and females in the

amygdala........................................................................................................................................59

Figure 17 Simplified diagram of the effect of Perinatal HFD on the general CORT/LPS

pathway..........................................................................................................................................66

List of Supplementary Tables

Supplementary Table 1

Transcriptional response to CORT challenge in the amygdala (post-hocs)................................101

Supplementary Table 2

Transcriptional response to CORT challenge in the hippocampus (post-hocs)...........................102

Supplementary Table 3

Transcriptional response to LPS challenge in the amygdala (post-hocs)....................................103

Supplementary Table 4

Transcriptional response to CORT/LPS challenge in the amygdala (post-hocs)........................104

xii

List of Abbreviations

11-β-HSD2 11-Beta hydroxysteroid dehydrogenase 2

ActB Beta-actin

ACTH Adrenocorticotropic hormone

ANOVA Analysis of variance

BMI Body mass index

CBG Corticosteroid-binding globulin

CHD Control House chow diet

ChIP Chromatin immunoprecipitation

CNS Central nervous system

Co-IP Co-immunoprecipitation

CORT Cortisol/corticosterone

CRP C-reactive protein

Fisher's PLSD Fisher's protected least significant difference

FKBP5 FK506 binding protein 5

Gapdh Glyceraldehyde 3-phosphate dehydrogenase

GC Glucocorticoid

xiii

HFD High-fat diet

HPA-axis Hypothalamic-pituitary-adrenal axis

IL-1β Interleukin-1 beta

LPS Lipopolysaccharide

LUMA Luminometric methylation assay

MAPK Mitogen-activated protein kinase

PI-3K Phosphoinositide 3-kinase

OD Optical density

PND Postnatal day

qRT-PCR/qPCR Quantitative real-time reverse transcriptase-polymerase chain reaction

rRNA Ribosomal RNA

Tet Ten-eleven translocation methylcytosine dioxygenase

TLR4 Toll-like receptor 4

TNF-α Tumor necrosis factor alpha

Ubc Ubiquitin C

Ywhaz 14-3-3 protein zeta/delta

Refer to Table 2 (pg.32) for genes analyzed with qPCR.

1

Chapter 1

1 Introduction

There is accumulating literature on the psychological effects that maternal obesity and

overnutrition, the predominant intake of high-fat foods, incurs on offspring of mammals,

including humans. At least two prevailing explanations exist to account for these effects: 1.

Differences in development of the hypothalamic-pituitary-adrenal (HPA)-axis during pre- and

post-(peri)natal periods of life, and 2. Inflammatory changes in the brain resulting from perinatal

exposure to saturated high fat diets (HFD) - the most commonly used paradigm to drive

overnutrition. These systems are not mutually exclusive: The HPA-axis regulates inflammation

through glucocorticoid and mineralocorticoid receptors (GR/MR), and cytokines, mediators of

inflammation, also regulate the HPA-axis. However, both can affect mental health through

separate mechanisms. Together, with the detrimental effects that overexposure to saturated fats

may have on brain development, makes offspring perinatally exposed manifest unique mental

and behavioral profiles during their lifespan compared to other early-life stressors or diet

manipulations. Engrained defects in activity of epigenetic regulators due to perinatal stress are

known to affect both systems, as they determine the extent that HPA-axis- and inflammatory-

related genes are transcribed. What is not clear, is how these genes may become differentially

regulated when the HPA-axis is activated by stressors experienced during adulthood. In this

study, we studied the GR/MR-mediated inflammatory pathway in limbic brain areas - regions

where the HPA-axis stress response is (dys)regulated by GR/MR. Here, we expected to see local

alterations in transcript abundance as a consequence of perinatal HFD exposure that would

indicate amplified stress and sickness-induced anxious and depressive-like behaviors, as well as

2

cognitive changes in rodents. Further, we expected to see diet-related differences in the activity

of epigenetic mediators, as well as on the regions of DNA that they target.

We will begin by reviewing literature that provides context for our findings - starting

with a brief overview regarding the societal impacts of maternal obesity and overnutrition.

Pertinent to the thesis, the remainder will be focused on the effects on anxiety- and depressive-

like behavioral responses in rodents, and how long-lasting alterations on the HPA-axis and

neuroinflammatory state could predispose individuals to these responses.

1.1 Effects of Maternal Obesity and Overnutrition on Offspring Physical and Mental

Health

68% of adults in the United States are now overweight or obese, including many women of

childbearing age (Flegal et al., 2010; Sullivan et al., 2010). Maternal obesity incurs health risks

for both the mother and their children, some of which manifest later in life. Indeed,

epidemiological studies often suggest that exposure to maternal obesity during early

development is a risk factor for cardiovascular disease, diabetes, and metabolic syndrome

(Godfrey Gluckman, and Hanson, 2010). Rodent and human studies further show heightened

incidences of eating disorders, including offspring preferences for fat-rich foods (Ong and

Muhlhausler, 2011; Rivera et al., 2015). Likely as a consequence, in addition the permanent

changes in metabolism, children of obese women tend to exhibit higher body mass index (BMI)

and body fat percentage (Simmons, 2008; Freeman, 2010). This suggests that the health risks can

be perpetuated through successive generations, calling into attention the need for more biological

research on how a mother's medical condition and dietary intake impacts development.

3

Studies in humans further suggest that offspring of obese mothers, and those that

consume high fat diets and drive weight gain during pregnancy, show an increased risk for

psychiatric ailments including autism, attention-deficit/hyperactivity disorder, schizophrenia,

cognitive impairments, anxiety, and depression (Sullivan et al., 2015; Krakowiak et al., 2012;

Grissom and Reyes, 2013; Rivera et al., 2015). The increased risk is in part a consequence of the

tendency for maternal obesity and weight gain to impair placental function and development, and

result in low or high birth weights, because the latter alone also poses risk for these diseases

(Grissom and Reyes, 2013; Rivera et al., 2015). In at least three studies, links between maternal

obesity and the development of anxiety and depression in children and adolescents have been

demonstrated: High maternal pre-pregnancy BMI has predicted reports of disrupted emotions in

children, such as fear and sadness, and increased internalizing behaviors which associate with

depression (Rodriguez, 2010; Van Lieshout et al., 2013). Another study correlated maternal

obesity with high or low birth weight, which coincided with the development of anxiety and

depression in adolescents (Coleman et al., 2012).

Several studies modelling maternal obesity with non-human primates as well as rodents,

through overnutrition with high fat diets (HFD), have also shown increased anxiety-like and fear

responses in the presence of novelty in adulthood during a variety of behavioral tests, which

frequently depended on the sex of the animals (Sullivan et al., 2010; Bilbo and Tsang, 2010;

Peleg-Raibstein, Luca, and Wolfrum, 2012; Sasaki et al., 2013). Furthermore, depressive-like

behavior has been reported in two studies with male offspring, where less time was spent

swimming and climbing in a Porsolt Forced Swim test (Can et al., 2012; Giriko et al., 2013).

These psychiatric illnesses in humans, and affect-related behavioral responses in rodents

are likely caused by malfunctions in at least two particular systems: The hypothalamic-pituitary-

4

adrenal (HPA)-axis, and inflammation of the central nervous system (CNS). Below, we describe

the comprehensive literature available on perinatal stressors in general, and what is known with

regards to maternal overnutrition, which we will establish as another type of 'stress'.

1.2 Perinatal Programming of the HPA-axis: Effect on Mental Health

Apart from maternal obesity or overnutrition, each of the psychiatric or behavioral

alterations have often been suggested to be due least in part by exposures to a variety of stressful

experiences which lead to abnormal brain development during the perinatal period. This includes

childhood maltreatment and trauma (Teicher and Samson, 2013; Shapero et al., 2014; Varese et

al., 2012) and maternal prenatal anxiety (O'Connor et al., 2002; van den Bergh et al., 2006), and

presence of stressors such as viral infection and adverse life events (Schmitt et al., 2014;

Markham and Koenig, 2011). Such experiences have been extensively shown to permanently

modify the activity of the developing hypothalamic-pituitary-adrenal (HPA) axis, which is a

major component of the neuroendocrine system that mediates response to stress (van Winkel et

al., 2008; Heim et al., 2000; MacKenzie et al., 2007; Weinstock, 2008). The HPA-axis regulates

processes including digestion, metabolism, behaviour, and the immune system according to

magnitude of perceived threat in the environment, in part by controlling the secretion of the

glucocorticoid, cortisol (corticosterone, in rodents) (Smith and Vale, 2006). Among other

upstream hormones such as corticotropin-releasing hormone (CRH), cortisol is circulated

throughout the body to signal physiological changes in tissues (Smith and Vale, 2006). For

instance, it increases levels of glucose in the blood to prepare for metabolic demands during

intense exercise (Robson et al., 1999).

5

Changes in gene expression of the receptors that bind cortisol, glucocorticoid receptors

(GRs) and mineralocorticoid receptors (MR), have often been reported following exposures to

pre and/or postnatal stress. In particular, the changes occur within the amygdala and

hippocampus, where both receptors are abundant, and specifically serve the purpose of

regulating the HPA-axis (Welberg and Seckl, 2000; Brunton, 2010; McGowan et al., 2009;

Weinstock, 2005). These brain regions are a part of the limbic system which regulates processes

including memory, emotion, behavior, and motivation. In the amygdala, the receptors are

involved in activating the HPA-axis, while negative feedback is initiated by receptors in the

hippocampus. Both are accomplished by enhancing or reducing the secretion of cortisol

respectively (Smith and Vale, 2006; Anacker et al., 2011; Qin et al., 2003). Since GR has a lower

affinity to cortisol than MR, it is only active during stress responses where levels increase, and

MR is saturated. On the other hand, MR is constitutively active (Korte, 2001). (Figure 1)

summarizes the relationships between each component of the HPA-axis.

6

Figure 1. Summary of the HPA-axis from Smith and Vale (2006). Neurons in the amygdala are

activated following exposures to stress. The signal is passed through projections innervating the

paraventricular nucleus of the hypothalamus, which releases corticotropin-releasing

factor/hormone (CRH). Receptors in the pituitary gland bind CRH, inducing the release of

adrenocorticotropic hormone (ACTH) into the bloodstream. The adrenal cortex binds ACTH,

causing the synthesis and release of cortisol/corticosterone (CORT), which then binds

glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) to carry out its systemic

metabolic, immune, behavioural, and cardiovascular functions. GR and MR in the hippocampus

bind cortisol to reduce the secretion of CORT upstream, thus gradually terminating the stress

response. Under high magnitude of stress, persistent circulation of CORT can further stimulate

the amygdala and maintain HPA-axis activity (Makino et al., 2002). Credit for the Diagram:

Hyman (2009).

7

1.2.1 Effects of GR and MR on Anxiety and Depression

A body of work has supported the role of limbic GR and MR in mediating anxiety and

depression, although the effects generally appears to depend on a number of factors, including

sex, brain region, and context. Limbic GR has frequently been connected to anxiety-like

behavior, typically increasing its expression (Welberg et al., 2000; Sarrazin et al., 2009;

Chmielarz et al., 2013; Durairaj and Koilmani, 2014). Its activity has also been shown to be

involved in the development and maintenance of major depression (Ströhle and Holsboer, 2003;

Anacker et al., 2011; Juruena et al., 2013) or depressive-like behavior (Chmielarz et al., 2013).

Several studies have suggested limbic MR to have a protective function against anxiety in

males, in the amygdala (Mitra et al., 2009), hippocampus (Herrero et al., 2006), and forebrain

(Lai et al., 2007; Rozeboom et al., 2007). On the other hand, one study suggests that

hippocampal MR, rather than GR, regulates the expression of anxiety in males (Smythe et al.,

1997). In contrast, a recent study suggests that forebrain MR, a part of the brain that includes the

limbic system, does not affect anxiety-like behavior in female mice (Kanatsou et al., 2015) -

conflicting with the study by (Rozeboom et al., 2007) which suggested that MR was also

protective for females.

A protective role of limbic MR also seems to exist for depression. In the 1990s, many

studies have shown that treatment of rats with antidepressant drugs increases the gene expression

and binding capacity of MR in limbic brain areas, including the hippocampus (Reul et al., 2000).

Hippocampal MR, but not GR has also been found decreased in patients with major depression

(Medina et al., 2013). Furthermore, a common haplotype of MR has been discovered to protect

against depression in women (Klok et al., 2011). Malfunctional activity of MR on HPA-axis

8

regulation have also been found in depressed patients (Juruena et al., 2013). In contrast, one

study suggests that patients with major depression maintain high functional activity of the MR

system (Young et al., 2003).

1.2.2 Overexposure to GCs alters GR and MR expression

Overall, limbic GR appears to increase anxiety and depressive behaviors, while limbic MR tends

to be protective. At least one way that altered gene expression of the receptors specifically occurs

during development is through excess fetal exposure to maternal or synthetic glucocorticoids

(GCs) (Welberg et al., 2000; Weinstock, 2005; Levitt et al., 1996), which has been shown to

occur in reaction to maternal prenatal anxiety (O'Donnell et al., 2011). More specifically,

increased exposure to GCs apparently increases mRNA levels of CRH in the amygdala, which

when individually administered during the last week of gestation, causes reductions in levels of

GR and MR in the hippocampus (Welberg et al., 2001). There is evidence to suggest that

cytokines activated by maternal overnutrition also results in fetal overexposure to GCs, which

will be discussed in more detail in Section 1.4 (Bellisario et al., 2015; Kossintseva et al., 2006).

This, with the fact that cytokines activate and regulate the HPA-axis (Kronfol, 2003), makes

maternal overnutrition another form of perinatal stress. Therefore, perinatal stressors each cause

similar mental health outcomes, in part because of how they increase fetal exposure to GCs.

1.3 Mediation of CORT- and LPS-induced Inflammatory Changes Through GR and MR

In addition to regulating the HPA-axis, GR mediates the expression of pro- and anti-

inflammatory molecules depending on the level and sensitivity to CORT - thereby contributing

to the role of the HPA-axis in controlling immune responses. Some studies also suggest the

involvement of MR, where it seems to potentiate inflammation in peripheral and central tissue

9

(Duan et al., 2013; Changtong et al., 2012). There is extensive evidence to indicate that central

and peripheral inflammation can result in psychiatric illnesses and anxiety- and depressive-like

behaviors, which is reviewed on Section 1.4. Therefore, one way that GR and MR modulates

anxiety and depression, among others, is through modifications of inflammatory state.

1.3.1 The General Immunosuppressive Action of GCs.

The involvement of GR in mediating immune responses has been well-documented (Sorrells et

al., 2009; Rhen and Cidlowski, 2005) . In most instances, it serves to suppress the activity of a

key pro-inflammatory transcription factor, Nuclear Factor Kappa Light Chain Enhancer of

Activated B cells (NFkB), upon recognition of an immune stressor. Classically,

lipopolysaccharide (LPS), a component in the cell membranes of gram-negative bacteria, is used

to activate NFkB. It binds to toll-like receptor 4 (TLR4) on the host cell membrane, and signals

Mitogen-activated protein kinase/Phosphoinositide 3-kinase (MAPK/PI-3K) pathways to activate

NFkB. It does this by causing Nuclear factor of kappa light polypeptide gene enhancer in B-

cells inhibitor, alpha (IkBa) to dissociate from NFkB, which is an anti-inflammatory protein that

inhibits its activity. NFkB is then capable of translocating to the nucleus to activate the

transcription of pro-inflammatory cytokines necessary for initiating an inflammatory response,

one of which is Interleukin-6 (IL-6) (Libermann and Baltimore, 1990).

Over time, the GC CORT is secreted, dissociates from corticosteroid-binding globulin

(CBG), and enters the cell. In the cytoplasm, it can either bind to MR, GR, or 11β-HSD, which

will degrade CORT. If it binds and activates GR, this causes the release of co-chaperones from

GR, including heat shock proteins 70 and 90, and FK506 binding protein 5 (FKBP5), while

expression of the latter is induced by GR over time to desensitize receptors to CORT (Binder,

10

2009). Activated GR forms a dimerized complex with CORT and translocates to the nucleus,

where it activates the expression of anti-inflammatory molecules, including IkBa, and MAP

kinase phosphatase-1 (MKP-1), which is a key molecule that is necessary to ameliorate the

immune response. The molecules translocate to the nucleus, where IkBa rebinds NFkB, and

MKP-1 inhibits the MAPK pathway. This mechanism of action is referred to as transactivation.

GR additionally suppresses inflammation by preventing NFkB from binding to its consensus

sites, kB RE. This indirect mechanism of action is known as transrepression. NFkB also begins

to activate the expression of anti-inflammatory cytokines such as Interleukin-10 (IL-10), which

inhibits NFkB (Saraiva and O'Garra, 2010; Driessler et al., 2004). A simplified diagram of this

process is illustrated in Figure 2.

11

Figure 2. Simplified diagram of the general CORT/LPS pathway. In the presence of LPS,

transcription factor NFkB is activated, allowing it to translocate to the nucleus to induce gene

expression of IL-6 and other pro-inflammatory cytokines. Later in the immune response, CORT

is secreted, and binds GR in the cytoplasm. GR is activated, and inhibits NFkB activity through

transactivation of anti-inflammatory molecules, IkBa and MKP-1, and direct transrepression of

NFkB. NFkB also increases expression of anti-inflammatory cytokine, IL-10 over time.

It is important to note, however, that basal CORT levels are needed to mount the initial

immune response against LPS, and that secretion of CORT stimulates the immune response to

acute, low-to-moderate physiological stress. This could be from the action of MR, but has not

been tested extensively. One study has indicated that in microglia, the resident immune cells in

the brain, MR potentiates the activation of NFkB, and expression of pro-inflammatory cytokines,

IL-6 and TNF-α, after treatments with low-to-moderate concentrations of CORT - supporting

this idea (Changtong et al., 2012). Therefore, the effect of GCs is only immunosuppressive when

it is: (1) secreted/administered after LPS, and (2) when they are administered chronically, or with

acute high-doses. ‘Acute’ refers to single-time stressors or injections of CORT, while ‘chronic’

refers to repeated stressors or injections given over a period of weeks or months, In fact, if either

acute or chronic CORT is administered prior to LPS, it facilitates the cell's pro-inflammatory

response. This also holds true in the central nervous system (CNS), but not for all regions of the

brain (Sorrells et al., 2009).

1.3.2 Inflammatory actions of GCs in the Central Nervous System (CNS).

In the CNS, acute stressors also stimulate inflammation, in one way by increasing NFkB activity.

In addition, higher basal CORT levels are associated with a greater accumulation of pro-

12

inflammatory mediators even in the absence of stress - likely via MR. The LPS-induced pro-

inflammatory actions of GCs in the CNS are known to at least in part be mediated by GR. In

spite of reductions in protein levels, GR is necessary to increase LPS-induced NFkB activation in

the hippocampus and cortex - regions abundant in GR, but not in the hypothalamus after acute or

chronic doses of CORT (Frank et al., 2009; Munhoz et al., 2006; de Pablos et al., 2006). As in

the periphery, CORT suppresses the immune response if given after LPS. One way that CORT is

known to mediate pro-inflammatory responses in the CNS is through GR-mediated increases in

proliferation and activation of microglia, the resident macrophage-like immune cells of the brain

(Nair and Bonneau, 2006; Frank et al., 2006), and another is through decreases in expression of

MKP-1 and IkBa (Munhoz et al., 2010). The reason GCs can sometimes activate NFkB in the

CNS, and not other times, is because it is protective in neurons (e.g. when facilitating

hippocampal synaptic plasticity; Albensi and Mattson, 2000), while being damaging to

microglia.

It must be noted, however, that GCs are not always pro-inflammatory in GR-abundant

brain regions. Reductions in NFkB signaling have also been found in the mouse hippocampus

following exposures to chronic psychosocial stress (Feldker et al., 2006). Thus, chronic stress

seems to be immunosuppressive in the CNS in the absence of an immune challenge, as in the

periphery. Acute, high-doses of CORT could also mimic the effects of chronic stress, as

observed by Mitra and Sapolsky (2008). In their study, they observed morphological changes

and inductions of anxiety of similar magnitude to those from chronically stressed male rats.

Similar indications have been made by Dhabhar (2000). Below, we begin to describe the

psychiatric and behavioral effects that microglial activity and inflammation has on the human

and rodent brain – with and without the involvement of GR and MR.

13

1.4. Neuroinflammation, GR, and Mental Disorders: Implications for Maternal

Overnutrition

When an individual or animal contracts a sickness, or is under high stress, what often occurs is

the development of depressive-like symptoms, such as anhedonia (reduced ability to experience

pleasure), reductions in food and water intake, reduced exploration, social, and physical activity.

These behavioral changes can be replicated by injections with pro-inflammatory cytokines,

indicating that it is not the pathogen itself, but the host immune response that causes these

symptoms (Najjar et al., 2013). Beginning in the periphery, many autoimmune illnesses and

medical conditions associated with chronic inflammation, such as systemic lupus erythematosus,

sepsis, obesity, diabetes, rheumatoid arthritis, or chronic fatigue syndrome, are often comorbid

with, or increase the risks for depression (Najjar et al., 2013; Zjaja, 2013; Afari and Buchward,

2003). In lupus and sepsis, the psychiatric symptoms may additionally include anxiety and

psychotic disturbances (Postal et al., 2011; Zjaja, 2013). Furthermore, patients with major

depressive disorder can be distinguished from healthy controls by immunological biomarkers in

their serum, including elevated plasma concentrations of pro-inflammatory cytokine, IL-6,

relative to anti-inflammatory cytokine, IL-10 - which indicates the net state of inflammation

(Dhabhar et al., 2009; Fredericks et al., 2009; Voorhees et al., 2013). Severe psychiatric illnesses

such as schizophrenia, have also been associated with increased serum levels of inflammatory

molecules in human birth cohorts (Ratnayake et al., 2013), having prior hospitalization for

serious infection, and having an autoimmune disease (Benros et al., 2011). As previously

mentioned in Section 1.2, perinatal exposures to maternal, or fetal infection can result in the

development of a milieu of psychiatric disorders or affect-related behaviors (Bilbo and Schwarz,

14

2009; Schmitt et al., 2014; Markham and Koenig, 2011), thereby providing evidence of a partial

involvement of cytokines in mediating psychopathology throughout the lifespan.

There are three processes by which circulation of cytokines in the periphery could

proceed to affect the limbic system (i.e. amygdala and hippocampus, which regulate affect and

affect-related behavior) within the CNS. The first is through cytokine receptors that line the

vagus nerve, which extends from the brainstem to multiple organs, including the esophagus,

heart, lungs, and gastrointestinal tract (Berthoud and Neuhuber, 2000). Second, "glymphatic"

vessels have also recently been discovered to provide immune cells from cerebrospinal fluid a

passage within, and out of the brain (Louveau et al., 2015). Third is a well-characterized passage

through the blood, where cytokines pass through the median eminence of the hypothalamus,

where the blood-brain-barrier is relatively weak (Kronfol, 2003). Here, they bind to receptors

including TLR4, and induce the production of soluble mediators that can cross into the brain and

activate microglial cells, which secrete cytokines through pathways including the NFkB system.

1.4.1 How Cytokines in the CNS Promote Psychopathology

The activation of microglia is crucial for the development of sickness behaviors during infection:

Selectively blocking inflammatory activity in the CNS can block these behaviour changes (Maier

and Watkins, 2012). Cytokines in the brain can modulate affect and behavior in several ways.

First, cytokines within the brain are exclusively involved in stimulating noradrenergic and

serotonergic systems, increasing the synthesis of tryptophan, and activating the HPA-axis by

increasing CORT circulation - all of which mediate affect and affect-related behavioral processes

(Kronfol, 2003). Further, since the brain is particularly vulnerable to oxidative stress, which is

provoked by neuroinflammation, the anti-inflammatory action of microglia are important for

15

maintaining the integrity of neural tissue and the blood-brain-barrier - both which are also

reported in psychiatric illnesses (Najjar et al., 2013). Similarly, depression has been suggested to

be caused at least in part by impairments in synaptic and structural plasticity due to elevations in

cytokine levels (Datson et al., 2013). Finally, cytokines also play a crucial role in the brain

during development, as they mediate the migration of neuronal and glial cells, differentiation,

and synaptic maturation (Nawa and Takei, 2006), while microglia are involved in processes such

as angiogenesis, axonal growth, and apoptosis. Thereby, one could imagine how the microglia in

the CNS can become sensitized to repeated or severe exposures to infections, or other forms of

stress that activate GR. During adulthood, this could enable transient sickness behaviors to

manifest as psychiatric illnesses. On the other hand, exposures during the perinatal period could

increase susceptibility to developing these illnesses throughout life.

There is extensive literature on the effects of cytokine expression in the developing

hippocampus during particular time-points, and its resultant positive and negative impacts on

memory, learning, and affect during adulthood, which has been reviewed by Bilbo and Schwarz

(2009). In essence, early immune challenges "prime" microglia to be more constitutively active,

pro-inflammatory, and reactive to stress during adulthood - either impairing or facilitating

performances during learning tasks, for example. Pertaining to maternal overnutrition, we will

now describe how perinatal exposures can alter cytokine profiles in the rodent brain, how this

can cause alterations in HPA-axis activity, and collectively affect behaviour and responses to

stress.

16

1.4.2 Caveats to Consider with Maternal Overnutrition Studies

Before we proceed, it must be noted that studies on maternal overnutrition tend to vary in the

type of diet used, and the duration of exposure. Most incorporate diet manipulations throughout

the perinatal period, and use diets that are high in fat. High fat diet (HFD), then, will be used

interchangeably with overnutrition. The composition of fats can differ between saturated,

unsaturated, and trans fats, and have varying proportions of protein and carbohydrate content.

'Cafeteria' diets are also sometimes used. Through the rest of the thesis, the focus will be on diets

high in saturated fat, the most common fat used to drive overnutrition.

1.4.3 Effects of Perinatal HFD on Inflammation

Potentially, what makes maternal saturated HFD unique compared to other perinatal stressors is

the ability of saturated fats to cross the blood-brain-barrier as well as the placental barrier

(Bolton and Bilbo, 2014). The implications of this process is two-fold: (1) saturated fats will be

the primary fat type integrated into the fetal brain, which is predominantly composed of fats, and

affect development, and (2) saturated (and trans) fats can access and bind to TLR4 within the

brain and dictate the distribution of local cytokines (Milanski et al., 2009). Because, in both

humans and rodents, the proportion of saturated fatty acids taken into the brain peak, then

continue decreasing through perinatal development to favour incorporation of other fat types

(Svennerholm and Vanier, 1978; Bourre et al., 1979), brain development could become restricted

by the deficits of other types of fats included in the diet. In humans, there is also evidence that

the mother's inflammatory state from ingesting saturated fats can be transmitted to the offspring

through releases of pro-inflammatory cytokines, which can cross into the fetal component

(Zaretsky et al. ,2004), as well as through the breast milk during postnatal development and

17

further activate TLR4 (Fagerås et al., 1999). Therefore, much like in the presence of infection,

HFD acts as an early-life 'challenge' that induces neuroinflammation in the developing fetus, and

sets the immunological template to reacting to future infections or stressful events.

In support of this, maternal HFD-exposed rodent offspring display increased circulating

levels of pro-inflammatory markers C-reactive protein (CRP) and IL-6 during birth, which

resemble those apparent after exposures to maternal infection (Williamson et al., 2011). In

humans, the restricted delivery of nutrients to the fetus (placental insufficiency) that often

happens as a result of maternal obesity also tends to be accompanied by pro-inflammatory

cytokine responses in the placenta (Wang et al., 2003; Bartha et al., 2003) – recalling that growth

restriction tends to be accompanied by future psychiatric illnesses (Section 1.1). During

adulthood, rodent models of placental insufficiency exhibit anxiety-like behavior and reduced

expressions of GABA and serotonin receptors in the hippocampus - indicating altered activity of

cytokines within the brain (Mikaelsson et al., 2013). Indeed, another maternal HFD rodent study

found increased brain levels of IL-1β, TNF-α, and microglial activation in female offspring

which associated with increased anxiety and decreased sociability (i.e. depression). Most

interestingly, replacing the maternal diets with standard chow during lactation was able to

alleviate these effects (Kang et al., 2014). Similarly, perinatal HFD males display heightened

levels of IL-6, microglial activation, and oxidative stress in the brain, which associated with

cognitive decline in the Morris water maze spatial learning task (White et al., 2009). In

summary, maternal HFD directly increases neuroinflammation, and indirectly by incurring

placental insufficiency - resulting in anxious- and depressive-like behavior and cognitive deficits

in learning tasks during adulthood.

18

1.4.4 Effects of Perinatal HFD on the HPA-axis as a Consequence of Inflammation.

Pro-inflammatory cytokines secreted due to exposures to saturated fats can also decrease the

activity of 11-β-HSD2, an enzyme that inactivates maternal GCs before it crosses the fetal

component (Kossintseva et al., 2006). This results in increased fetal exposure to CORT, and

should mimic the effects of physiological/psychological stressors on the expression of limbic GR

and MR. The resultant impact on GR and MR would then further impact the activity of microglia

and distribution of cytokines, and drive affect and behavioral abnormalities experienced with or

without stressful life experiences. Hence, saturated fats separately expose offspring to impaired

brain development, GR/MR, and pro-inflammatory cytokine expression during development,

which all have downstream consequences on affect and affect-related behaviors that at times

overlap - making it a distinct from other perinatal stressors.

To our knowledge, the effect of perinatal HFD on the expression of GR and MR in adults

has only been examined in our recent study (Sasaki et al., 2013), although some literature exists

on chronic consumptions of HFD during adulthood - with similar results (Sasaki et al., 2015).

We found that exposure to maternal saturated HFD in rodents increases the basal transcript levels

of GR and/or MR in the amygdala mostly for females (recalling that it activates the HPA-axis),

and such changes are associated with alterations in abundance of target pro- and anti-

inflammatory molecules, including those shown in (Figure 2). In HFD females, subtle decreases

in anti-inflammatory transcript abundance were also apparent in the hippocampus. Coinciding

with the changes in transcripts were enhanced anxiety responses to the open field, elevated plus

maze, and light-dark transition tests. A hyperactive HPA-axis response was also apparent in the

HFD females, such that CORT levels remained elevated above the control chow diet (CHD)

females during the recovery period from restraint stress. This was not observed in the males. Our

19

study further suggested that maternal HFD may preferentially alter GR/MR, and inflammatory

responses in the amygdala compared to the hippocampus, bringing into attention the need for

more studies on maternal HFD in the former limbic region.

Epigenetic processes, which dynamically mediate the expression of genes depending on

cues taken from the environment, is known to be permanently altered by maternal HFD. Systems

affected by perinatal stress-induced changes in epigenetic regulation include those of the HPA-

axis, and those that mediate immune responses. Therefore, alterations of the two systems are in

fact manifestations of underlying differences in epigenetic regulation - making this an additional

layer that has to be considered. We will now discuss how DNA methylation, one means of

epigenetic regulation, causes permanent changes in expression of the related genes in response to

perinatal stressors, including maternal HFD. Pertinent to this thesis, we will also describe the

involvement of epigenetic mechanisms in responding to stress challenges during adulthood. In

essence, some studies indicate that impaired stress responses can occur from exposures to prior

stressors, and that this could be due in part by dysregulated expression of molecules that mediate

epigenetic processes.

1.5 Role of Epigenetic Regulation in Incurring Lifelong Effects of Maternal Overnutrition

To begin, a number of commonly-cited studies examining lifelong effects of postnatal

experiences such as quality of maternal care in rodents, or exposures to childhood abuse and

neglect in humans have found changes in expression of limbic GR and MR to be the result of

alterations in DNA methylation at their promoters (Weaver et al., 2004; Weaver et al., 2007;

McGowan et al., 2009). DNA methylation is an epigenetic mechanism generally indicated to

regulate gene expression by either directly preventing the binding of repressive or activating

20

components of the transcription machinery, or signaling to configure the surrounding chromatin

into making the DNA more or less accessible to the transcription machinery (Jones, 2012;

Fagiolini et al., 2009). Altered promoter methylation of GR has also been observed with prenatal

exposure to maternal depression or chronic variable stress in humans and rodents respectively

(Oberlander et al., 2008; Mueller and Bale, 2008). In the liver, reduced DNA methylation of GR,

and increased expression has been reported following maternal protein-restricted diets during

pregnancy (Lillycrop et al., 2007). Similarly,

in response to increased quality and quantity

of maternal care, the DNA methylation of

IL-10, an anti-inflammatory cytokine that

also regulates the HPA-axis (Smith et al.,

1999), has been found reduced specifically in

the microglia, and related to increases in

expression (Schwarz et al., 2011).

In addition to the alterations in

methylation state resulting from perinatal

stress, the activity and expression of

molecules which regulate DNA methylation

has been suggested to become impaired,

particularly in response to perinatal diet.

McKee and colleagues (2014) have found

that maternal saturated HFD (60% to 20%

protein and carbohydrates) results in reduced DNA methyltransferase (DNMT) enzymatic

Box 1. Regulators of DNA methylation.

DNMTs are the enzymes responsible for

appending methylation marks to DNA.

MeCP2 is a chromatin-associated protein that

binds methylated DNA to sterically block

binding of the transcription machinery, and

interact with histone deacetylases (HDACs)

to alter the chromatin state (Theisen et al.,

2013). The isoforms of DNMTs, maintenance

methyltransferases DNMT1 which sustain

existing methylation, and de novo

methyltransferases DNMT3a and DNMT3b

which add novel methyl groups (Jeltsch and

Jurkowska, 2014), are known to interact with

one another and MeCP2 to carry out their

specific activities (Fatemi et al., 2002;

Hseieh, 2005; Kimura and Shiota, 2003). On

the other hand, Gadd45b and Tet are factors

and enzymes invovled in actively

demethylating DNA (Niehrs and Schäfer,

2012; Kohli and Zhang, 2013).

21

activity in the prefrontal cortex that coincided with decreased interactions of DNMT1 isoforms

with Methyl-CpG-binding Protein 2 (MeCP2) (Box 1). The impaired DNMT activity co-

occurred with global hypomethylation of promoters, and overall increased gene expression.

Unexpectedly, the gene and protein levels of DNMT1 were overexpressed in this study,

suggesting a compensatory mechanism for the reduced function. Other studies indicate

reductions in global and gene-specific DNA methylation, including GR, in response to

nutritional deficits during gestation (Doherty et al., 2000; Lillycrop et al., 2008), associating with

decreased mRNA levels of DNMT1 in newborn (Pham et al., 2003) and adult rodents (Lillycrop

et al., 2007). The other epigenetic mechanisms, histone modifications and non-coding

microRNAs, have also been found to be affected by exposures to nutritional factors during

perinatal development (Canani et al., 2011).

1.6 Role of Epigenetic Regulation in Responding to Stress

1.6.1 Gene-specific Functions

Epigenetic mechanisms are known to play a role in mediating the transcriptional response to

stress. When bound to CORT, activated GR normally regulates transcription by associating with

epigenetic regulators, with one example involving interactions with DNMTs, MeCP2, and

HDAC1 in the repression of hypothalamic CRH. This serves as a means to initiate negative

feedback on the HPA-axis (Sharma et al., 2013). To terminate the stress response, GR is also

downregulated over time in order to reduce sensitivity to CORT (Chiba et al., 2012; Gehring et

al., 1984), and this is also mediated epigenetically. In one mechanism, activated GR binds to its

own negative consensus GR-binding site (nGRE) at a distal regulatory region, then recruits

HDAC3 to the promoter-proximal region of GR to form a repression complex and inhibit

22

transcription (Ramamoorthy and Cidlowski, 2013). Pertaining most to this thesis, MeCP2 is

known to repress the expression of downstream targets in response to CORT and Tumour

Necrosis Factor alpha (TNF-α), an immunological stressor. This includes FKBP5 and IL-6

respectively, in the microglia (Cronk et al., 2015).

1.6.2 Mediation of Global Changes

The involvement of epigenetic processes in responding to GCs is by no means limited to a select

number of genes. Rapid changes in histone marks occur in the hippocampus during responses to

acute stressors such as forced swim and novelty tests, which would result in genome-wise

changes in gene transcription through altered global repatterning of chromatin state (Datson et

al., 2013). During late stages of fetal development, a natural GC surge occurs in order to

facilitate the maturation of several organ systems through genome-wide changes in gene

expression (Crudo et al., 2012). This surge has been observed to be accompanied by global

changes in CpG methylation in a number of peripheral tissues that persist into adulthood (Crudo

et al., 2012). CpG refers to the dinucleotide sequence where DNA methylation most often

occurs, and serves as the binding site for MeCP2 (Jones and Takai, 2001). They are commonly

found in promoters of mammalian genes, and tend to be found unmethylated when the genes are

expressed. The converse is true for those residing within gene coding regions. This forms the

basis for a number of epigenetic techniques developed to approximate the amount of DNA

methylation in particular regions or genomic samples (Suzuki and Adrian, 2008).

1.7 The Effects of Prior Experiences on Responses to Stress

Since early-life stressors such as maternal HFD appear to have a 'priming' effect on epigenetic

regulators such that its enzymatic activity is permanently altered, then its activity and interaction

23

with GR may also be impaired during responses to stress. It is currently unclear how

inflammatory pathways in the brain, thereby affect and behaviour, would be affected by the

presence of a perceived threat after prior stress, because studies that examine its consequences

upon re-induction of stress have generally been scarce. Here, we review the existing literature on

the effects that prior chronic stress has on the HPA-axis activity, inflammation, epigenetic

regulation, and transcriptional response to acute stressors.

1.7.1 Studies inspecting HPA-axis Activity

Previous exposures to chronic social isolation have been reported to lead to decreased secretion

of CORT and increased NFkB signaling in response to acute restraint stress in the liver

(Djordjevic et al., 2010). Prior exposures to a single prolonged stress paradigm have also been

shown to impair the regulation of of GR and MR mRNA expression during recovery to an acute

stressor in the hippocampus (Liberzon et al., 1999): Initially, both downregulate expression, but

MR remains downregulated 1-2 weeks later, while GR reaches higher than prestress levels. On

the other hand, animals that did not display altered negative feedback responses after the initial

prolonged stressor were able to normalize GR/MR levels.

Similarly, there is evidence that maternally separated neonatal rats can maintain adequate

or enhanced HPA-axis activity during baseline, while displaying prolonged activity in response

to acute stressors coinciding with differential GR/MR gene expression in the hippocampus (Ladd

et al., 2004). Comparing between rats with shorter periods of maternal separation, ACTH and

CORT levels were sustained for longer times in response to an airpuff startle stimulus. And, in

response to a dexamethasone suppression test which is used to gauge negative feedback

responses to exogenous CORT, ACTH and CORT levels returned to baseline levels sooner. It

24

was found that hippocampal MR mRNA density had initially been enhanced in these rats, while

hippocampal GR was reduced - explaining the impairments in negative feedback. This study

suggested that some effects of prior chronic stress may only be apparent when GR is activated,

such as if the chronic stressor is of moderate intensity.

In contrast to these psychological stressors, rats exposed to bacterial infections early in

life showed faster recovery from depressive symptoms induced by CORT, and blunted HPA-axis

responses (i.e. reduced serum CORT) during and after the stressor - although the depressive

symptoms were more pronounced (Bilbo et al., 2008). Hence, there is preliminary evidence to

support that perinatal stress affects the responsiveness of the HPA-axis during further activation,

but that the outcome appears to vary according to the initial type of stress. Therefore, it is again

pertinent to note the distinctiveness of maternal saturated HFD from other types of early-life

stressors.

1.7.2 Studies examining Transcriptional and Epigenetic effects

Along similar lines as with Ladd et al (2004), effects of prior chronic stress in the rodent

hippocampus have only been unmasked after GR was activated with acute, high-dose

corticosterone (CORT) injections (Datson et al., 2013). Compared to animals with no previous

exposure to chronic restraint stress, about 60% of genes affected by CORT were differentially

expressed (p < 0.05). This included several genes involved in chromatin structure and epigenetic

processes, which supported that the changes in expression may have been partially causing the

differential transcriptional response to CORT. In this study, genes from the TNF-α/NFkB

signaling pathway were exclusively affected by the presence of prior restraint after administration

of acute CORT - noting that the population isolated was predominantly neuronal cells. The

25

directionality, however, was not specified. Connections to epigenetic regulation have also been

implicated in a recent study using restraint stress and LPS in the stress paradigm. It was

discovered that the gene expression of a number of epigenetic mediators failed to upregulate in

the hypothalamus of maternal HFD adult rats, which showed overall reduced transcriptional

responses in a panel of 26 genes (Grissom et al., 2015).

1.7.3 Other studies with Inflammatory Pathways

To our knowledge, there has only been one study with maternal HFD (saturated and trans fat)

that examined changes in cytokine expression in response to LPS by Bilbo and Tsang (2010).

They discovered that the protein levels of pro-inflammatory cytokine, IL-1β in the rodent adult

hippocampus were strikingly increased both basally, and post-LPS, and this co-occurred with

higher basal and LPS-induced microglial activation - suggesting a chronic priming effect. The

HFD, non-challenged offspring also displayed increased anxiety, but enhanced performance in a

spatial learning task, the Morris Water Maze. This was perhaps due to the elevations of IL-β,

which is crucial for normal learning (Williamson et al., 2011).

1.8 Research Hypotheses

To our knowledge, the majority of research on maternal HFD, including epigenetic studies, has

focused on the consequences on peripheral tissues. These studies also tend to exclude females

offspring due to the effects of the stage of estrus on measurements such as behavior. Moreover,

measurements have typically been taken with unstressed animals, while more studies have begun

to unmask effects of prior chronic stress during GR activation (Ladd et al., 2004; Datson et al.,

2013). Given the prior evidence that genes such as GR and MR would be differentially expressed

(Sasaki et al., 2013), we were interested in examining effects of maternal saturated HFD on the

26

limbic brain areas of both sexes after 'challenges' with different types of stressors: Acute stress

with high-dose CORT, and/or bacterial infection with injections of LPS. In this study, we chose

to focus on differences in transcript levels of components from the established inflammatory

pathway targeted by GR and MR, as outlined on Section 1.3 because of our interest in

psychiatric illnesses, and the fact that we had seen diet-related differences in our previous study

(Sasaki et al., 2013). We expected diet-related differences to exist in the amygdala and

hippocampus, given that they are the regions where the HPA-axis is (dys)regulated by GR/MR,

and where local inflammatory changes would result in anxiety/depressive behaviors.

We hypothesized that:

1) Perinatal HFD-exposed animals will display aberrant transcriptional inflammatory

responses to each challenge. In response to the acute, high-dose of CORT used, we

predicted its immunosuppressive properties would become enhanced due to GR/MR

being permanently conditioned by the prior exposure to chronic stress. In contrast, we

expect animals to show heightened pro-inflammatory responses to LPS, as observed by

Bilbo and Tsang (2010), due to the perinatally sensitized state of existing microglia. With

simultaneous injections of CORT/LPS, we then expected that the perinatal overexposure

to CORT would result in similar pro-inflammatory changes as when CORT is given

hours before LPS. If this is the case, we should observe increased transcript abundance of

CD11b, a marker for microglia.

2) Given the study by Bilbo and colleagues (2008), and the fact that anxious and depressive

behaviors are already apparent in non-stressed animals, we expected perinatal HFD-

exposed rats to also carry a higher propensity for reacting to stress with anxious- and

depressive-behavior than CHD rats. Therefore, we predicted that the expression of genes

27

associated with anxiety and depression would become elevated in the HFD groups. This

includes CRH in the amygdala, associated with anxiety (McGill et al., 2006; Samaco et

al., 2012); and MKP-1 and IL-6/IL-10, associated with depression (Duric et al. 2010,

Dhabhar et al., 2009; Fredericks et al., 2009; Voorhees et al., 2013).

3) We hypothesized that DNMT transcript levels and activity will be altered in perinatal

HFD-exposed animals to partially explain the differential transcript responses we might

observe. Since the maternal HFD study by McKee et al. (2014) found reduced DNMT

activity in non-stressed animals, with apparent compensatory increases in DNMT1

expression, we also expect to see this with our HFD groups before and during the stress

challenges.

4) If DNMT activity is reduced, then global or gene-specific differences in proportion of

DNA methylation should also accompany the transcript differences. Given that DNMTs

methylate DNA, we expected global CpG methylation to overall be reduced in limbic

brain areas. In response to CORT, which at least up- and down-regulates an even number

of genes in the hippocampus (Datson et al., 2013), the net outcome of reduced DNMT

activity should be further reductions in global CpG methylation. .

28

Chapter 2

2 Materials & Methods

2.1 Animals

Adult male and female Long Evans rats (7 week) used were obtained from Charles River Canada

(St. Constant, QC). Rats were housed in same-sex pairs, and maintained on a 12:12-h light-dark

cycle (lights on from 7:00 am - 7:00 pm) with ad libitum access to food and water. Experimental

protocols were approved by the Local Animal Care Committee at the University of Toronto,

Scarborough, and were in accordance with the guidelines of the Canadian Council on Animal

Care.

2.2 Diets

Female breeders were placed on one of two diets: A HFD containing 60% saturated fat, 20%

carbohydrate, and 20% protein (n = 15), or control house chow diet (CHD) with 58%

carbohydrate, 28.5% protein, 13.5% saturated fat (n = 14). The 5.24-kcal/g HFD was obtained

from Research Diets, Inc. (New Brunswick, NJ: cat. no. D12492), and the 3.02-kcal/g CHD was

obtained from Purina Lab Diets (St. Louis, MO: cat. no. 5001). Similar formulations have

previously been compared to examine diet-induced obesity (El-Haschimi et al., 2000; de Souza

et al., 2005; Dunn and Bale, 2009; Tamashiro et al., 2009; Purcell et al., 2011). The females

remained on the diet for 4 weeks prior to mating, and throughout pregnancy and lactation. Upon

weaning at Postnatal day (PND) 21, offspring were maintained on CHD throughout adulthood. A

timeline for the diet regimen is illustrated in Figure 3.

29

Figure 3. Perinatal-HFD regimen used in this study. The animals were injected with CORT,

LPS, or CORT/LPS, and brain regions were collected 3 hours later for downstream applications

such as qPCR for gene expression i.e. transcript level analysis.

2.3 Subjects and general procedures

Female breeders were housed individually after mating, with no significant differences in litter

size or sex ratio among the diet groups. Except for weighing during weekly cage changes,

offspring remained undisturbed until weaning at PND 21, when they were housed in same-sex

pairs. Prior to the drug challenges, body weights of the offspring were measured (n = 167).

2.4 Drug challenges

During PND 90, the adult offspring were injected with either an acute, subcutaneous dose of

CORT dissolved in propylene glycol (10 mg/kg, Sigma-Aldrich, cat. no. 27840), a low

intraperitoneal dose of LPS from Escherichia coli dissolved in saline (25 ug/kg, Sigma-Aldrich,

cat no. L2630), or a simultaneous high-dose CORT/LPS injection (10 mg/kg, 50 ug/kg) (n = 6

per diet and sex). To determine diet x drug effects, a subset evenly divided by sex and diet were

left non-injected (n = 24). The dose of CORT has been shown to result in CORT levels

comparable to that of several hours of high physiological stress (Stein-Behrens et al., 1994), and

is sufficient to induce anxiety and dendritic hypertrophy on the amygdala of similar magnitude to

30

those from chronic treatments (Mitra and Sapolsky, 2008). The 25yg/kg dose used for LPS has

been used in an extensive body of studies on neonatal infection cited in Williamson et al. (2011),

where cognitive impairments and altered cytokine profiles were observed in the hippocampus

after exposures to further stress and immune challenges. For the 50 ug/kg dose of LPS, enhanced

hippocampal microglial activation and pro-inflammatory cytokine expression was previously

observed in maternal HFD-exposed groups (Bilbo and Tsang, 2010). All of the conditions used

in this project are outlined in Table 1.

Table 1. Number of adult offspring in each challenge condition. Each cell includes an even

number of male and female rats (CORT: mg/kg, LPS: ug/kg). Control offspring were left

undisturbed until sacrifice. The conditions that will be discussed are highlighted.

CORT (s.c.) LPS (i.p.) CORT+ LPS

Dosage Control 5 10 Control 25 50 Control 5, 25 10, 50

CHD 4 12 12 4 12 12 4 12 12

HFD 4 12 12 4 12 12 4 12 12

Total 56 56 56

2.5 Brain tissue preparation

The adult offspring were sacrificed by CO2 inhalation followed by decapitation. Brains were

rapidly dissected, flash-frozen with isopentane, and stored at -80ºC. Using stereotaxic

coordinates, the entire dorsal hippocampus and amygdala was extracted under a Leica CM3050

cryostat. Tissues were homogenized with TRIzol Reagent (Invitrogen, Carlsbad, CA, USA), with

31

1 ml per 50-100 mg of tissue, and RNA, DNA, and proteins were separately extracted. RNA

were first extracted with RNeasy Mini Plus Kits (Qiagen, Valencia, CA, USA), then DNA and

proteins were simultaneously extracted with a manufacturer's protocol for TRIzol isolations

optimized for purity and concentration. Quantification/quality assessments were performed with

a Nanodrop ND-2000C spectrophotometer according to the manufacturers' protocol. The DC

Protein Assay (Bio-Rad, Hercules, CA, USA) was used for the protein quantifications.

2.6 Analyses of transcript abundance by quantitative real-time reverse transcriptase-polymerase

chain reaction (qRT-PCR)

Transcript abundance of genes involved in glucocorticoid signaling, mediating inflammation,

and epigenetic regulation were quantified by StepOne Plus real-time PCR using Fast SYBR

Green PCR master mix (Applied Biosystems, Life Technologies, Carlsbad, CA, USA) and by

relative normalization against the levels of each of five housekeeping genes to compare between

groups (Gapdh, ActB, Ubc, Ywhaz, and rRNA). Table 2 lists the analyzed genes with brief

descriptions. geNorm software selected Gapdh, Ywhaz, and ActB as the three most stably

expressed between the diet and drug conditions. ActB was selected for the analysis due to its

high similarities with results using the geometric mean of the three genes and its common usage

in other studies of HFD. To enable comparisons between runs, transcript abundance of each gene

was quantified by the standard curve method, where the same cDNA mixture of known

concentrations was run with every sample per tissue. All the reactions were performed in

triplicate.

32

Table 2. List of genes analyzed for transcript abundance with qPCR.

Functional Group Gene Abbrev. Description

Glucocorticoid

(GC) Signaling

Glucocorticoid

Receptor

GR Binds GCs to regulate genes involved in stress-induced

HPA-axis activity (Heegde et al., 2015)

Mineralocorticoid

Receptor

MR Binds GCs to regulate genes involved in basal- and

stress-induced HPA-axis activity (Heegde et al., 2015)

Corticotropin-

releasing hormone

CRH Stimulates secretion of ACTH in the pituitary to produce

cortisol/corticosterone (Smith and Vale, 2006)

Pro-inflammatory Cluster of

differentiation

molecule 11b

CD11b Microglial marker (Bilbo and Tsang, 2010).

Nuclear Factor

Kappa Light Chain

Enhancer of

Activated B cells

NFkB Key inflammatory transcription factor (Nelson et al.,

2003).

Interleukin-6 IL-6 Pro-inflammatory cytokine regulated by NFkB

(Libermann and Baltimore, 1990)

Interleukin-

6/Interleukin-10

IL-6/IL-

10

Marker of inflammation and depression (Dhabhar et al.,

2009; Fredericks et al., 2009; Voorhees et al., 2013).

Anti-

inflammatory

Insulin-like growth

factor 1

IGF-1 Neuroprotective; behaves as anti-inflammatory cytokine

in the brain (Mangiola et al., 2015; Bake et al., 2014)

Nuclear factor of

kappa light

polypeptide gene

enhancer in B-cells

inhibitor, alpha

IkBa Inhibits NFkB until, and during activation (Nelson et al.,

2003).

MAP kinase

phosphatase-1

MKP-1 Crucial for resolving inflammation (Vandevyver et al.,

2012); marker for depression (Duric et al. 2010)

Interleukin-10 IL-10 Anti-inflammatory cytokine against, and regulated by,

NFkB (Driessler et al., 2004; Saraiva and O'Garra, 2010).

Epigenetic

regulators

DNA

methyltransferase

1

DNMT1 Maintenance methyltransferase (Jeltsch and Jurkowska,

2014)

DNA

methyltransferase

3a

DNMT3

a

de novo methytransferase (Jeltsch and Jurkowska, 2014)

DNA

methyltransferase

3b

DNMT3

b

de novo methytransferase (Jeltsch and Jurkowska, 2014)

Methyl CpG

binding protein 2

MeCP2 Binds methylated CpG sequences to regulate

transcription (Theisen et al., 2013)

Growth Arrest And

DNA-Damage-

Inducible, Beta

Gadd45

b

Stress and activity-induced DNA demethylation to

enable neurotypical development (Kigar et al., 2015)

33

2.7 Primer specifications

The primers used for qRT-PCR were purchased from Qiagen, or designed using sequence

information from GenBank at the National Center for Biotechnology Information (NCBI;

www.ncbi.nlm.nih.gov), as shown on (Table 3).

Table 3. Primer sequences and catalogue numbers for genes analyzed with qPCR.

Gene Sequence

Forward Primer Reverse Primer

Ubc 5'-CAC CAA GAA GGT CAA ACA GGA A-3' 5'-AAG ACA CCT CCC CAT CAA ACC-3'

Gapdh 5'-ACA TCA AAT GGG GTG ATG CT-3' 5'-GTG GTT CAC ACC CAT CAC AA-3'

ActB 5'-TTT GAG ACC TTC AAC ACC CC-3' 5'-ATA GCT CTT CTC CAG GGA GG-3'

18S rRNA 5'-ATG GTA GTC GCC GTG CCT A-3' 5'-CTG CTG CCT TCC TTG GAT G-3'

Ywhaz 5'-TTG AGC AGA AGA CGG AAG GT-3' 5'-GAA GCA TTG GGG ATC AAG AA-3'

MR 5'-GGC AGC TGC AAA GTC TTC TT-3' 5'-GAC AGT TCT TTC GCC GAA TC-3'

CRH 5'-CTC TCT GGA TCT CAC CTT CCA C-3' 5'-CTA AAT GCA GAA TCG TTT TGG C-3'

CD11b 5'-GAA GCC TTG GCG TGT GAT AG-3' 5'-GAG CAG TTT GTT CCC AAG GG-3'

IGF-1 5'-GCT CTT CAG TTC GTG TGT GG-3' 5'-TGA GTC TTG GGC ATG TCA GT-3'

IkBa 5'-CAG GAT TCT GCA GGT CCA CT-3' 5'-TGG AGC ACT TGG TGA CTT TG-3'

MKP-1 5'-GCT CCA CTC AAG TCT TCT TCC TCC AA-3' 5'-TGG ACT GTT TGC TGC ACA GCT

CAG-3'

IL-10 5'-AGA AGG ACC AGC TGG ACA ACA C-3' 5'-GTC GCA GCT GTA TCC AGA GGG-3'

DNMT1 5'-ACC TAC CAC GCC GAC AT-3' 5'-AGG TCC TCT CCG TAC TCC A-3'

DNMT3a 5'-ACG CCA AAG AAG TGT CTG CT-3' 5'-CTT TGC CCT GCT TTA TGG AG-3'

DNMT3b 5'-GAT GTG ACA CCT AAG AGC AGC AGT AC-3' 5'-CAA ACT CCT TGT CAT CCT GAT ACT

CA-3'

MeCP2 5'-CAA ACA GCG ACG TTC CAT CA-3' 5'-TGT TTA AGC TTT CGC GTC CAA-3'

Gadd45b 5'-GCT GGC CAT AGA CGA AGA AG-3' 5'-GCC TGA TAC CCT GAC GAT GT-3'

GR RT2 quantitative-PCR (qPCR) Primer Assay (PPR52805B, SA biosciences, Qiagen)

NFkB RT2 qPCR Primer Assay (PPR42746A, SA biosciences, Qiagen)

IL-6 RT2 quantitative-PCR (qPCR) Primer Assay (PPR06483B, SA biosciences, Qiagen, Valencia,

CA, USA)

34

2.8 Measurement of DNMT enzyme activity and abundance in the amygdala

To quantify the total enzymatic activity of DNMTs, the EpiQuikTM DNMT Activity/Inhibition

Assay Ultra Kit (Fluorometric) was used (Epigentek, Farmingdale, NY, USA). In triplicates, 5-

10 ug of proteins were added into wells stably coated with an oligonucleotide sequence optimally

designed to be methylated by all active DNMT isoforms in the presence of the substrate, S-

Adenosyl methionine. Anti-5-methylcytosine was added to capture the amount of methylated

DNA, and fluorescence signals generated from a dye-bound secondary antibody were detected to

calculate total enzymatic activity in units of RFU/h/mg with the formula, DNMT Activity =

(Sample RFU - Blank RFU)/(Protein Amount (µg) x hours incubation) x 1000. Using EpiQuikTM

DNMT1 Assay Kit (Epigentek, Farmingdale, NY, USA), a similar principle was used to quantify

the amount of DNMT1 protein colorimetrically using anti-DNMT1 in place of 5mC, with the

formula DNMT1 (ng/mg protein) = (Sample OD - Blank OD)/(Slope x Protein Amount (µg)) x

1000. OD = Optical density.

2.9 Global CpG methylation analysis in the amygdala with Luminometric methylation assay

(LUMA)

LUMA is a restriction enzyme-based method of estimating the global proportion of CpG

methylation using the isoschizomers, HpaII and MspI which both target CCGG sequences, with

the difference being that HpaII cannot cleave the sequence when the internal cytosine is

methylated. Methylation most often occurs on cytosines of CpG dinucleotides (Jones and Takai,

2001), hence it would be an approximate representation of the total amount of methylation

present in a sample of genomic DNA. EcoRI is added to each reaction to control for the

efficiency of each digest, allowing for the cleavage ratios to only capture the methylation

35

difference. A combination of protocols from Karimi et al.(2006)(Original), Pilsner et al.(2010),

and Crudo et al. (2012) were used for this procedure. Briefly, 300 ng of genomic DNA was

added to two duplicate reactions containing 5U each of Hpa/II EcoRI or MspI/EcoRI in Tango

Buffer. Distilled water was included for a total volume of 20 ul. The samples were incubated at

37ºC for 4 h, then heat-inactivated at 80ºC for 20 min. 20 ul of Pyrosequencing annealing buffer

(Qiagen) was added to each reaction, and the samples were transferred onto 96-well

Pyrosequencing plates. The nucleotide dispensation order from Pilsner et al. (2010) was used,

and Pyrosequencing was conducted with PyroMark MD (Qiagen). Peak heights of nucleotide

incorporation were used to calculate % genomic DNA methylation using the formula: 1-

[(HpaII(First G after addition of C)/EcoRI(First T after addition of A))/MspI(G)/EcoRI(T))] x

100, where CG and AT represent the overhangs left behind after each digest.

2.10 Statistical analyses

The weights of pups were analyzed by 2 x 2 day-by-diet repeated measures analysis of variance

(ANOVA), and adults, by 2 x 2 diet by sex ANOVA on SPSS (IBM Corporation, Armonk, NY,

USA). Gene expression, LUMA, and DNMT data were analyzed by 2 x 2 diet by drug ANOVA

on Statview (SAS institute, Cary NC) for each sex. All significant ANOVA tests were followed

by Fisher's PLSD post hoc comparisons. Spearman's Correlations for DNMT activity were

performed on SPSS. IL-6/IL-10 ratios were normalized against non-injected transcript levels in

order to compare between each drug challenge. This was done by subtracting values from the

mean expression levels of non-injected HFD and CHD groups. For the qPCR analyses, all results

used for interpretation are presented as bar graphs.

36

Chapter 3

3 Results

3.1 Perinatal-HFD offspring display differences in body weight throughout development

The Prenatal-HFD exposed offspring displayed lower gestational body weights during PND 1 (p

< 0.05), while maintaining higher body weights after postnatal HFD-exposure by PND 17 (p <

0.01) [Day by diet interaction: F(1,24) = 11.903, p < 0.0001, Figure 4a]. Although the adults had

been fed CHD since PND 21, the perinatal HFD-exposed rats maintained higher body weights

than the CHD group prior to the drug challenges at PND 90 [F(1,165) = 82.455, p < 0.0001], with

males weighing more than females on average [Diet by sex interaction: F(1,165) = 7.587, p < 0.01,

Figure 4b].

Figure 4. Body weights of offspring, as (a) pups from PND 1-21, Bar****: p < 0.0001 Diet by

day interaction; *, **: p < 0.05, p < 0.01 post-hoc comparison within day, and (b) adults prior to

drug challenges, Bar****: p < 0.0001 Main effect of diet; ****: p < 0.0001 Main effect of sex.

a) b)

37

3.2 Perinatal-HFD exposed animals display exacerbated anti-inflammatory transcriptional

responses to CORT challenge in the amygdala

In response to CORT, pro- and anti-inflammatory transcript levels significantly increased for

both diet groups (Table 4; Post-hocs: Supplementary Table 1). Both diet groups for males

exhibited higher relative abundance of IL-6 and IL-6/IL-10 ratios post-CORT (Figure 5a).

Increases for anti-inflammatory molecules, IkBa and MKP-1 were significantly higher for the

HFD males than the CHD males [Diet by drug interaction: IkBa: F(1,23) = 12.020 , p < 0.01, MKP-

1: F(1,23) = 7.977 , p < 0.01; Figure 5b]. This coincided with a maintenance of higher levels of GC

signaling molecules than the CHD males [GR: F(1,23) = 4.909 , p < 0.05, CRH: Diet by drug

interaction: F(1,23) = 3.295 , p < 0.1], which significantly downregulated GR (p < 0.01) while HFD

males slightly increased CRH expression (p < 0.1) (Figure 5c).

The females displayed sparse changes in transcript abundance when responding to CORT (Table

4b). HFD females significantly increase the amount of pro- and anti-inflammatory transcript

levels [IL-6/IL-10: F(1,23) = 5.687 , p < 0.05, IkBa: F(1,23) = 9.065, p < 0.01; Figure 6a,b], while

decreasing in abundance of GR (p < 0.05) (Figure 6c).

38

a) Pro-inflammatory Genes

b) Anti-inflammatory Genes

b) GC-signaling Genes

39

Figure 5. Transcript level data for CORT-challenged males in the amygdala. (a) Pro-

inflammatory transcripts are increased for both diet groups. (b) Perinatal HFD-exposed males

maintain higher transcription of anti-inflammatory genes in response to CORT, and (b)

transcription of GC-signaling genes are sustained and enhanced for Perinatal HFD-exposed

males. Bar**, #: p < 0.01, p < 0.1 post-hoc comparison within diet, **, *: p < 0.01, p < 0.05

post-hoc comparison within drug,

a) Pro-inflammatory Genes b) Anti-inflammatory Genes

c) GC-signaling genes

Figure 6. Transcript level data for CORT-challenged females in the amygdala. (a) Perinatal

HFD-exposed females increase pro- and b) anti-inflammatory transcript abundance in response

40

to CORT, and (b) abundance of GR is reduced for Perinatal HFD-exposed females. Bar*: p <

0.05 post-hoc comparison within diet.

Table 4. Transcriptional response to CORT challenge in the amygdala. 2 x 2 ANOVAs of qPCR

gene expression data separated by (a) males and (b) females. P-values < 0.05 are highlighted.

a) Males

b) Females

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT CHD CORT F P F P F P

GC signalling GR 1.3 1.246 0.996 1.181 0.305 0.5872 2.414 0.1368 1.003 0.3291

MR 1.155 1.37 1.168 1.236 1.748 0.2027 0.317 0.5802 0.47 0.5019

CRH 0.511 0.542 0.601 0.495 0.147 0.7061 0.047 0.8314 0.479 0.4973

Pro-inflammatory CD11b 0.679 0.811 0.514 0.695 1.812 0.195 1.469 0.2412 0.046 0.8324

NFkB 1.027 0.92 0.891 0.822 1.203 0.2865 2.117 0.162 0.056 0.8153

IL-6 1.255 1.594 1.98 1.594 0.004 0.9493 1.003 0.3306 1.002 0.3308

IL-6/IL-10 0.916 1.053 1.25 1.206 0.207 0.6548 5.687 0.283 0.787 0.3867

Anti-inflammatory IGF-1 0.829 0.913 0.912 0.866 0.028 0.8683 0.027 0.8716 0.342 0.5656

IkBa 0.443 0.447 0.763 0.611 0.839 0.3711 9.065 0.0072 0.932 0.3466

MKP-1 0.574 0.602 0.727 0.608 0.421 0.5248 1.277 0.2732 1.075 0.3135

IL-10 1.568 1.538 1.573 1.311 0.287 0.5988 0.165 0.6893 0.182 0.6745

Epigenetic regulation DNMT1 0.506 0.531 0.694 0.59 0.179 0.6771 1.742 0.2026 0.482 0.496

DNMT3a 0.572 0.647 0.514 0.609 1.251 0.2781 0.403 0.5335 0.018 0.8937

DNMT3b 0.473 0.558 0.274 0.401 1.321 0.2647 3.691 0.0699 0.051 0.823

MeCP2 0.523 0.531 0.524 0.482 0.048 0.8279 0.098 0.7573 0.104 0.7508

Gadd45b 0.397 0.441 0.49 0.464 0.021 0.887 0.849 0.3683 0.322 0.577

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT CHD CORT F P F P F P

GC signalling GR 1.132 1.035 0.942 0.703 4.909 0.0385 11.796 0.0026 0.875 0.3609

MR 1.095 1.394 1.043 1.021 2.3 0.145 5.459 0.03 3.104 0.0934

CRH 0.411 0.513 0.649 0.34 0.834 0.3721 0.083 0.7767 3.295 0.0845

Pro-inflammatory CD11b 0.498 0.748 0.773 0.501 0.006 0.9401 0.01 0.9197 3.485 0.0767

NFkB 0.825 0.859 0.735 0.677 0.022 0.8829 3.075 0.0948 0.35 0.5608

IL-6 0.683 0.625 1.869 1.547 0.436 0.5165 13.37 0.0016 0.209 0.6523

IL-6/IL-10 0.285 0.245 1.084 1.183 0.038 0.8474 32.65 < 0.0001 0.209 0.6527

Anti-inflammatory IGF-1 0.868 0.946 0.879 0.642 0.805 0.3802 2.716 0.115 3.116 0.0928

IkBa 0.332 0.313 1.097 0.78 15.432 0.0008 207.037 < 0.0001 12.02 0.0024

MKP-1 0.449 0.494 0.997 0.673 4.54 0.0457 31 < 0.0001 7.977 0.0105

IL-10 2.319 2.763 1.906 1.497 0.001 0.9702 3.286 0.0849 0.846 0.3687

Epigenetic regulation DNMT1 0.632 0.731 0.827 0.469 2.781 0.111 0.186 0.6706 8.607 0.0082

DNMT3a 0.871 1.09 0.581 0.454 0.219 0.645 21.908 0.0001 3.063 0.0954

DNMT3b 0.405 0.571 0.45 0.436 0.376 0.5466 0.134 0.7177 0.525 0.4769

MeCP2 0.584 0.599 0.723 0.407 3.786 0.0659 0.118 0.7352 4.586 0.0447

Gadd45b 0.367 0.406 0.589 0.381 1.865 0.1872 2.539 0.1268 3.997 0.0594

41

3.3 Perinatal-HFD exposure incurs similar, but sex-dependent transcriptional responses to

CORT in the hippocampus

In the hippocampus, pro- and anti-inflammatory transcript levels increased in response to CORT

for all groups except the CHD females, where they were all reduced (Table 5; Post-hocs:

Supplementary Table 2). The transcript levels of these genes and GR were initially higher than

the HFD female groups, which had not been observed in the amygdala. Following CORT-

injection, the diet-differences in GR, CD11b, and NFkB levels were thereby abolished (Figure

8a,c). On the other hand, the abundance of anti-inflammatory genes, IkBa and MKP-1 increased

for HFD females to the extent that it exceeded those of CHD females (Figure 8b). A trend of

increased IL-6, and IL-6/IL-10 ratios was also only apparent for HFD-challenged females (p <

0.1, Supplementary Table 2). For both sexes, elevations in anti-inflammatory transcription

associated with either a maintenance of higher GR levels relative to CHD groups, or a lack of

reduction in response to CORT [Males: F(1,23) = 4.655, p < 0.05, Females: Diet by drug

interaction: F(1,23) = 6.457, p < 0.05, Figure 7c, 8c].

a) Pro-inflammatory Genes

42

b) Anti-inflammatory Genes

c) GC-signaling Genes

Figure 7. Transcript level data for CORT-challenged males in the hippocampus. (a) Pro-

inflammatory transcription is increased for both diet groups. (b) Perinatal HFD-exposed males

maintain higher levels of anti-inflammatory genes in response to CORT, and (b) levels of GC-

signaling genes are sustained for Perinatal HFD-exposed males. Bar****, **, *: p < 0.0001, p <

0.01, p < 0.05 post-hoc comparison within diet, *, #: p < 0.05, p < 0.1 post-hoc comparison

within drug.

43

a) Pro-inflammatory Genes

b) Anti-inflammatory Genes c) GC-signaling Genes

Figure 8. Transcript level data for CORT-challenged

females in the hippocampus. (a) Regular diet-exposed

females reduce transcription of pro-inflammatory

genes and (c) GR relative to Perinatal HFD levels in

response to CORT. (b) HFD-CORT females maintain

higher abundance of anti-inflammatory genes than

CHD-CORT. Bar**, *, #: p < 0.01, p < 0.05, p < 0.1

post-hoc comparison within diet, *, #: p < 0.05, p <

0.1 post-hoc comparison within drug.

44

Table 5. Transcriptional response to CORT challenge in the hippocampus. 2 x 2 ANOVAs of

qPCR gene expression data separated by (a) males and (b) females. P-values < 0.05 are

highlighted.

a) Males

b) Females

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT CHD CORT F P F P F P

GC signalling GR 1.688 1.534 1.62 0.961 4.655 0.0433 2.895 0.1044 1.792 0.1957

MR 1.633 1.49 2.09 1.134 4.837 0.0398 0.041 0.8418 2.653 0.119

Pro-inflammatory CD11b 1.283 1.424 1.559 1.206 0.188 0.6696 0.014 0.9066 1.026 0.3232

NFkB 1.082 0.896 1.286 0.864 4.067 0.0574 0.324 0.5757 0.616 0.4418

IL-6 1.347 0.745 3.01 4.04 0.213 0.6494 28.663 < 0.0001 3.108 0.094

IL-6/IL-10 0.633 0.237 1.291 1.327 0.524 0.478 12.419 0.0023 0.757 0.395

Anti-inflammatory IGF-1 1.199 1.188 2.02 1.486 1.039 0.3209 4.362 0.0505 0.95 0.3419

IkBa 0.504 0.458 2.245 1.128 7.459 0.0129 32.086 < 0.0001 6.333 0.0205

MKP-1 0.607 0.53 2.109 1.112 7.517 0.0126 28.303 < 0.0001 5.515 0.0292

IL-10 3.168 3.054 2.783 3.595 0.329 0.5733 0.016 0.8997 0.579 0.4562

Epigenetic regulation DNMT1 0.588 0.61 1.089 0.576 1.202 0.286 1.086 0.3098 1.423 0.2469

DNMT3a 0.674 0.649 0.799 0.555 1.059 0.3158 0.013 0.9093 0.703 0.4115

MeCP2 0.589 0.674 0.789 0.528 0.456 0.5071 0.043 0.8378 1.752 0.2006

Gadd45b 0.73 0.774 1.295 0.734 1.39 0.2523 1.427 0.2463 1.902 0.1831

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT CHD CORT F P F P F P

GC signalling GR 1.282 1.818 1.265 1.071 1.416 0.248 7.057 0.0152 6.457 0.0194

MR 1.032 1.245 1.018 0.985 0.751 0.3964 1.746 0.2013 1.411 0.2488

Pro-inflammatory CD11b 0.995 1.695 0.991 0.836 2.253 0.149 5.652 0.0275 5.534 0.029

NFkB 1.068 1.602 1.216 1.052 2.278 0.1468 2.673 0.1177 8.079 0.0101

IL-6 0.83 1.152 1.38 1.104 0.014 0.9067 1.693 0.2079 2.397 0.1373

IL-6/IL-10 0.424 0.806 1.262 0.635 0.23 0.637 1.71 0.2058 3.911 0.0619

Anti-inflammatory IGF-1 1.395 1.633 1.511 1.423 0.213 0.6492 0.082 0.7772 0.995 0.3305

IkBa 0.48 0.634 1.16 0.751 2.136 0.1594 20.98 0.0002 10.486 0.0041

MKP-1 0.439 0.662 0.806 0.514 0.136 0.716 1.374 0.2549 7.651 0.0119

IL-10 2.052 2.049 1.951 1.791 0.034 0.8564 0.164 0.69 0.032 0.8602

Epigenetic regulation DNMT1 1.573 1.709 0.851 0.647 0.014 0.9079 9.355 0.0062 0.341 0.5657

DNMT3a 1.681 1.672 0.685 0.56 0.046 0.8316 11.563 0.0028 0.034 0.8549

MeCP2 1.047 1.34 0.711 0.526 0.236 0.6326 26.153 < 0.0001 4.512 0.0463

Gadd45b 1.297 1.1 0.751 0.553 0.754 0.3955 5.778 0.026 2.12E-06 0.9989

45

3.4 Perinatal HFD-exposed animals display exacerbated anti-inflammatory transcriptional

responses to LPS challenge in the amygdala

Both pro- and anti-inflammatory transcript levels increased for LPS-challenged subjects,

excluding the CHD females (Table 6; Post-hocs: Supplementary Table 3). HFD males

additionally increased in MKP-1 [F(1,23) = 6.450 , p < 0.05, Figure 9a] while displaying more

significant increases in IkBa (p < 0.01) and less significant drops in IL-6/IL-10 ratios (p < 0.05).

HFD females increased IkBa transcript levels while abundance of anti-inflammatory genes were

unchanged for CHD females [F(1,23) = 7.205 , p < 0.05, Figure 10b]. Heightened pro-

inflammatory activity was further evidenced for the CHD females, with higher transcription of

NFkB [F(1,23) = 4.360, p < 0.05; Figure 10a]. Post-hoc analyses revealed this to only be

significant after injection with LPS (p < 0.1) (Figure 10a). Both CHD males and females

reduced MR and GR expression [MR: p < 0.1, GR: F(1,23) = 4.928, p < 0.05; Figure 9b, 10b].

a) Anti-inflammatory Genes b) GC-signaling Genes

Figure 9. Transcript level data for LPS-challenged males in the amygdala. (a) Perinatal HFD-

exposed males increase MKP-1 levels in response to LPS, and (b) levels of MR are sustained for

Perinatal HFD-exposed males. Bar *, #: p < 0.05, p < 0.1 post-hoc comparison within diet.

46

a) Pro-inflammatory Genes b) Anti-inflammatory Genes

b) GC-signaling Genes

Figure 10. Transcript level data for LPS-challenged females in the amygdala. (a) CHD-LPS

females sustain higher abundance of NFkB than HFD-LPS females, while (b) Perinatal HFD-

exposed females increase IkBa in response to LPS, and (c) levels of GR are sustained for

Perinatal HFD-exposed females. Bar *, #: p < 0.05, p < 0.1 post-hoc comparison within diet, *: p

< 0.05 post-hoc comparison within drug.

47

Table 6. Transcriptional response to LPS challenge in the amygdala.. 2 x 2 ANOVAs of qPCR

gene expression data separated by (a) males and (b) females. P-values < 0.05 are highlighted.

a) Males

b) Females

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD LPS CHD LPS F P F P F P

GC signalling GR 0.83 0.792 0.708 0.698 0.081 0.7795 1.66 0.2123 0.028 0.8695

MR 0.981 1.096 0.97 0.906 0.053 0.8209 0.801 0.3815 0.634 0.4532

Pro-inflammatory CD11b 0.849 1.417 1.094 0.986 1.72 0.2045 0.279 0.6034 3.723 0.068

NFkB 1.029 1.045 1.322 1.535 0.147 0.7055 1.724 0.204 0.11 0.7437

IL-6 0.417 0.495 2.031 1.988 0.001 0.9773 6.491 0.0192 0.01 0.9221

IL-6/IL-10 0.043 0.033 0.912 0.986 0.027 0.8711 22.377 0.0001 0.047 0.8305

Anti-inflammatory IGF-1 1.128 1.107 1.369 1.492 0.156 0.6973 5.851 0.0252 0.303 0.588

IkBa 0.342 0.379 0.678 0.724 0.266 0.6117 17.963 0.0004 0.003 0.9566

MKP-1 0.465 0.495 0.762 0.981 0.652 0.429 6.45 0.0195 0.375 0.5473

IL-10 9.359 15.054 2.322 1.984 0.898 0.3546 12.652 0.002 1.139 0.2986

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD LPS CHD LPS F P F P F P

GC signalling GR 0.704 0.823 0.565 0.594 0.809 0.3803 4.928 0.0395 0.297 0.5923

MR 0.944 1.055 1.123 1.366 2.201 0.1535 4.254 0.0524 0.305 0.5869

Pro-inflammatory CD11b 0.916 1.035 0.856 0.558 0.463 0.5042 4.163 0.0548 2.512 0.1287

NFkB 1.275 2.402 1.163 3.557 4.36 0.0498 0.383 0.5429 0.565 0.461

IL-6 0.365 0.281 1.573 2.892 0.76 0.3936 7.277 0.0139 0.981 0.3337

IL-6/IL-10 0.122 0.147 1.214 1.082 0.096 0.7602 34.875 < 0.0001 0.208 0.6529

Anti-inflammatory IGF-1 0.953 1.134 0.849 1.158 2.77 0.1116 0.074 0.7888 0.19 0.6673

IkBa 0.369 0.47 0.78 0.906 0.513 0.483 7.205 0.0151 0.006 0.9379

MKP-1 0.813 0.898 0.825 0.986 0.526 0.4778 0.087 0.7713 0.05 0.8258

IL-10 2.928 2.914 1.829 2.495 0.31 0.5836 1.685 0.209 0.337 0.5679

48

3.5 Perinatal-HFD exposed animals display altered pro-inflammatory transcriptional responses

to CORT/LPS challenge in the amygdala

When responding to CORT/LPS, the IL-6/IL-10 ratio increased higher than when injected with

the same dose of LPS alone, suggesting a synergistic effect of CORT. However, post-hoc

analyses suggest it is only significant for CHD groups [Males: F(1,22) = 4.715, p < 0.05, Females:

Diet by drug interaction: F(1,23) = 6.228, p < 0.05; Figure 11]. In this condition, HFD groups

reduce transcript levels of GR, and for females, the levels drop lower than the CHD group

(Table 7; Post-hocs: Supplementary Table 4). Coinciding with the lower levels of GR, MR,

and CRH is lower pro-inflammatory transcription for the HFD females (Figure 13). It is less

clear what is occurring with the HFD males, as higher NFkB levels are maintained alongside a

trend of reduced IL-6/IL-10 ratios (Figure 12a). In response to CORT/LPS, CHD males reduce

MR to levels equivalent to basal levels for HFD males (Figure 12b).

Figure 11. Increases in IL-6/IL-10 ratios between LPS and CORT/LPS. Bar*, #: p < 0.05, p <

0.1 post-hoc comparison within diet; #: p < 0.1 post-hoc comparison within drug. The increases

in IL-6/IL-10 ratios for all groups are significant (p < 0.01).

49

a) Pro-inflammatory Genes

b) GC-signaling Genes

Figure 12. Transcript level data for CORT/LPS-challenged males in the amygdala. (a) HFD-

CORT/LPS males sustain higher NFkB levels, but lower IL-6/IL-10 ratios than CHD-

CORT/LPS males. (b) Regular diet-exposed males reduce levels of MR to Perinatal HFD levels

in response to CORT/LPS. Bar***, **, *: p < 0.001, p < 0.01, p < 0.05 post-hoc comparison

within diet, *, #: p < 0.05, p < 0.1 post-hoc comparison within drug.

50

a) Pro -inflammatory genes b) GC signaling genes

Figure 13. Transcript level data for CORT/LPS-challenged females in the amygdala. (a)

CORT/LPS-induced pro-inflammatory signaling is dampened for Perinatal HFD-exposed

females, and (b) abundance of GC signaling genes are lower than for CHD females. Bar **, *: p

< 0.01, p < 0.05 post-hoc comparison within diet, **, *, #: p < 0.01, p < 0.05, p < 0.1 post-hoc

comparison within drug.

51

Table 7. Transcriptional response to CORT/LPS challenge in the amygdala. 2 x 2 ANOVAs of

qPCR gene expression data separated by (a) males and (b) females. P-values < 0.05 are

highlighted.

a) Males

b) Females

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT/LPS CHD CORT/LPS F P F P F P

GC signalling GR 1.595 1.628 1.027 0.926 0.091 0.7657 32.485 < 0.0001 0.359 0.5559

MR 1.73 2.297 1.42 1.148 0.882 0.3595 21.517 0.0002 7.104 0.0153

CRH 0.614 0.485 0.603 0.751 0.012 0.9143 2.33 0.1434 2.732 0.1148

Pro-inflammatory CD11b 1.057 1.047 0.891 0.927 0.004 0.9533 0.457 0.5071 0.012 0.9156

NFkB 0.622 0.656 0.409 0.322 0.208 0.6535 22.474 0.0001 1.117 0.3039

IL-6 0.392 0.366 1.461 1.214 0.857 0.3662 42.237 < 0.0001 0.56 0.4635

IL-6/IL-10 0.147 0.185 0.971 1.265 3.059 0.0973 100.509 < 0.0001 1.829 0.193

Anti-inflammatory IGF-1 1.231 1.46 0.782 0.697 0.298 0.5915 21.051 0.0002 1.4 0.2513

IkBa 0.373 0.378 0.694 0.563 1.648 0.2147 26.994 < 0.0001 1.937 0.1801

MKP-1 0.476 0.518 0.525 0.46 0.063 0.8043 0.01 0.9214 1.371 0.2561

IL-10 2.684 2.613 1.545 1.042 0.772 0.3907 17.266 0.0005 0.439 0.5156

Epigenetic regulation DNMT1 1.568 1.486 1.197 1.081 0.584 0.4539 9.007 0.0073 0.017 0.8972

DNMT3a 1.182 1.426 1.158 1.072 0.381 0.5445 2.207 0.1538 1.678 0.2107

DNMT3b 0.399 0.699 0.449 0.379 0.933 0.3463 1.275 0.2728 2.395 0.1382

MeCP2 0.77 0.82 0.873 0.82 0.001 0.9812 0.666 0.4245 0.692 0.4157

Gadd45b 1.295 1.027 1.162 1.386 0.022 0.8828 0.577 0.4562 2.708 0.1154

Mean relative abundance Diet Drug Diet x Drug

Function Gene HFD CTL CHD CTL HFD CORT/LPS CHD CORT/LPS F P F P F P

GC signalling GR 1.826 1.757 0.787 1.193 0.779 0.388 17.633 0.0004 1.549 0.2276

MR 2.203 2.4 1.16 1.493 1.263 0.2745 17.123 0.0005 0.083 0.7764

CRH 0.753 0.535 0.641 1.009 0.447 0.5114 2.588 0.1233 6.758 0.0171

Pro-inflammatoryCD11b 2.583 2.509 1.121 1.431 0.161 0.6921 18.534 0.0003 0.424 0.5222

NFkB 1.703 1.794 0.567 0.836 0.615 0.4421 20.705 0.0002 0.148 0.7042

IL-6 1.736 1.671 1.659 2.989 1.164 0.2934 1.12 0.3025 1.415 0.2481

IL-6/IL-10 0.686 0.639 1.796 4.693 4.359 0.0498 14.315 0.0012 4.651 0.0434

Anti-inflammatoryIGF-1 1.387 1.313 0.852 0.887 0.006 0.9385 3.765 0.0666 0.049 0.8278

IkBa 0.655 0.726 0.777 0.802 0.304 0.5873 1.283 0.2708 0.07 0.7945

MKP-1 0.762 0.757 0.4 0.508 0.225 0.6406 7.94 0.0106 0.27 0.6088

IL-10 2.55 2.639 1.096 0.743 0.18 0.6762 28.941 < 0.0001 0.503 0.4863

Epigenetic regulationDNMT1 1.558 1.572 0.93 1.312 1.111 0.3044 5.577 0.0284 0.957 0.3396

DNMT3a 1.257 1.335 0.788 1.072 0.891 0.3565 3.638 0.0709 0.287 0.5981

DNMT3b 1.065 0.903 0.534 0.9 0.353 0.5591 2.405 0.1366 2.352 0.1408

MeCP2 0.794 0.876 0.859 1.012 1.538 0.2293 1.129 0.3006 0.142 0.7106

Gadd45b 0.978 1.591 1.262 1.485 4.085 0.0568 0.185 0.6716 0.885 0.3582

52

3.6 Diet-dependent transcriptional responses cluster with transcript levels of epigenetic

regulators in the amygdala

Next, we examined the relative abundance of the DNMT isoforms, MeCP2, and Gadd45b in

CORT- and CORT/LPS-challenged individuals, where most of the diet, diet x drug, and sex

effects had been observed (Tables 4, 7; Supplementary Tables 1, 4). Post-hoc analyses revealed

that the directionality of the diet x drug interactions tended to resemble, or ‘cluster’ with those of

GR, MR, and CRH, and the inflammatory genes exhibiting immunosuppressive activity. In

particular, this can be observed for the CORT-injected males, and the CORT/LPS-injected

females in the amygdala (Table 8a,b; Supplementary Table 1a, 4b). For both, the levels of

DNMT1 were commonly clustered.

Table 8. Transcriptional response to CORT and CORT/LPS in the amygdala grouped by post-

hoc comparisons within diet and drug. Changes in transcript levels of epigenetic mediators

cluster with those impacted by Perinatal HFD for (a) CORT-challenged males, and (b)

CORT/LPS-challenged females. # p < 0.1, * p < 0.05, ** p < 0.01, n.s. not significant. Shaded

boxes: Diet x Drug interaction p < 0.05.

a) CORT-injected Males

Function Gene Effect of CORT within diet group Effect of diet within drug group

GC signalling GR CHD CTL > CHD CORT ** HFD CORT > CHD CORT *

MR CHD CTL > CHD CORT * HFD CTL < CHD CTL #

CRH HFD CTL < HFD CORT # HFD CORT > CHD CORT *

Anti-inflammatory IGF-1 CHD CTL > CHD CORT # HFD CORT > CHD CORT #

IkBa CTL < CORT, HFD + CHD ** HFD CORT > CHD CORT **

MKP-1 HFD CTL < HFD CORT ** HFD CORT > CHD CORT *

Epigenetic regulation DNMT1 CHD CTL > CHD CORT * HFD CORT > CHD CORT **

MeCP2 CHD CTL > CHD CORT # HFD CORT > CHD CORT *

Gadd45b HFD CTL < HFD CORT # HFD CORT > CHD CORT #

53

b) CORT/LPS-injected females

3.7 DNMT activity reduces in response to CORT and CORT/LPS for Perinatal HFD-exposed

males, but not CORT/LPS females in the amygdala

Our transcript analyses of the DNMTs provoked us to examine what may be occurring with the

enzymes on the molecular level. We observed that in response to CORT, HFD males showed

robust decreases in DNMT activity (p < 0.05), while it slightly increased for CHD males (p <

0.1) (Figure 14a). For the CORT/LPS condition, HFD males once again decreased DNMT

activity (p < 0.05), while it remained unchanged for CHD (Figure 14b). Females did not show

changes in DNMT activity in response to CORT/LPS (Figure14c). Spearman's Correlations

revealed that DNMT activity correlates with DNMT1 transcript levels, and not DNMT3a or

DNMT3b in both CORT and CORT/LPS-injected males. DNMT1 strongly correlates negatively

to activity for CORT (rho = -0.578, p < 0.01), while a positive trend is apparent in response to

CORT/LPS (rho = 0.364, p < 0.1) (Table 9). In both instances, DNMT1 also strongly correlates

to DNMT3a transcript levels (CORT: rho = 0.487, p < 0.05, CORT+LPS: rho = 0.786, p < 0.01).

There were non-significant increases in protein levels in CORT-injected HFD and CHD males,

Function Gene Effect of CORT/LPS within diet group Effect of diet within drug group

GC signalling GR CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS **

MR CTL > CORT/LPS, CHD **, HFD * HFD CORT/LPS < CHD CORT/LPS **

CRH CHD CTL < CHD CORT/LPS ** HFD CORT/LPS < CHD CORT/LPS **

Pro-inflammatory CD11b CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS *

NFkB CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS #

IL-6/IL-10 CTL < CORT/LPS, HFD + CHD ** HFD CORT/LPS < CHD CORT/LPS #

Epigenetic regulation DNMT1 HFD CTL > HFD CORT/LPS * HFD CORT/LPS < CHD CORT/LPS *

DNMT3a HFD CTL > HFD CORT/LPS # HFD CORT/LPS < CHD CORT/LPS *

DNMT3b HFD CTL > HFD CORT/LPS * n.s.

Gadd45b n.s. HFD CTL < CHD CTL #

54

which did not correlate with either transcript levels or DNMT activity (mRNA: rho = 0.015, p =

0.946, DNMT activity: rho = 0.109, p = 0.629) (Figure 14d).

0

500000

1000000

1500000

2000000

2500000

3000000

3500000

4000000

4500000

Non-injected CORT+LPS

DN

MT

Act

ivit

y (

RF

U/h

/mg

)

HFD-Males

CHD-Males

0

200

400

600

800

1000

1200

DN

MT

1 (

ng

/mg

Pro

tein

)

HFD

CHD

0

500000

1000000

1500000

2000000

2500000

3000000

3500000

Non-injected CORT

DN

MT

Act

ivit

y (

RF

U/h

/mg

)

HFD-Males

CHD-Males

0

1000000

2000000

3000000

4000000

5000000

6000000

7000000

Non-injected CORT+LPS

DN

MT

Act

ivit

y (

RF

U/h

/mg

)

HFD-Females

CHD-Females

**

____*____

a) CORT-injected males

b) CORT/LPS-injected males

c) CORT/LPS-injected females

d) DNMT1 protein levels

Figure 14. DNMT Activity and Protein

Assays. a) Total DNMT activity for

CORT-challenged males. **: p < 0.01

post-hoc diet comparisons within drug.

Post-hoc drug comparisons within diet

(not shown): CHD p < 0.1, HFD p <

0.05. b) Total DNMT activity for

CORT/LPS-challenged males. Bar *: p

< 0.05 post-hoc drug comparisons

within diet (HFD). c) Total DNMT

activity for CORT/LPS-challenged

females (n.s.). d) DNMT1 protein levels

for CORT-challenged males (n.s.).

55

Table 9. Spearman's Correlation Tests Between DNMT transcript abundance and Total DNMT

activity. DNMT activity correlates with DNMT1, which correlates with DNMT3a. *: p < 0.05, **:

p < 0.01.

CORT Males

DNMT1 DNMT3a DNMT3b DNMT_Activity

Spearman's

rho

DNMT1 Correlation

Coefficient

1 .487* 0.276 -.578**

Sig. (2-tailed) . 0.016 0.192 0.004

N 24 24 24 23

DNMT3a Correlation

Coefficient

.487* 1 0.223 -0.229

Sig. (2-tailed) 0.016 . 0.296 0.293

N 24 24 24 23

DNMT3b Correlation

Coefficient

0.276 0.223 1 -0.173

Sig. (2-tailed) 0.192 0.296 . 0.43

N 24 24 24 23

DNMT_Activity Correlation

Coefficient

-.578** -0.229 -0.173 1

Sig. (2-tailed) 0.004 0.293 0.43 .

N 23 23 23 23

CORT+LPS Males

DNMT1 DNMT3a DNMT3b DNMT_Activity

Spearman's

rho

DNMT1 Correlation

Coefficient

1 .786** 0.122 0.364

Sig. (2-tailed) . 0 0.581 0.096

N 23 23 23 22

DNMT3a Correlation

Coefficient

.786** 1 0.052 0.011

Sig. (2-tailed) 0 . 0.812 0.962

N 23 23 23 22

DNMT3b Correlation

Coefficient

0.122 0.052 1 -0.064

Sig. (2-tailed) 0.581 0.812 . 0.778

N 23 23 23 22

DNMT_Activity Correlation

Coefficient

0.364 0.011 -0.064 1

Sig. (2-tailed) 0.096 0.962 0.778 .

N 22 22 22 22

56

3.8 DNMT activity correlates with global CpG methylation in CORT-injected male groups in the

amygdala

Given that strong diet-dependent differences in DNMT activity were evident for the CORT-

challenged males in the amygdala, we examined whether this would be reflect genome-wide

changes in methylation using the LUMA technique. It was observed that CHD males sustained

significantly lower methylation levels than HFD males after responding to CORT [F(1,19) =

10.455, p < 0.01, Figure 15a). Correlating the DNMT activity of CORT-challenged groups

revealed significances with highly linear R2 values (HFD: rho = 0.829 , p < 0.05, R2 = 0.9635,

CHD: rho = 1.000, p < 0.01, R2 = 0.9902; Figure 15b). Therefore, DNMTs at least in part

mediates transcriptional responses to CORT that are apparent at an inter-individual level.

Figure 15. Global CpG methylation measured by LUMA in the amygdala of CORT-injected

males. (a) HFD males maintain higher % methylation than CHD in response to CORT, **: p

<0.01 post-hoc comparison within drug. (b) DNMT activity correlates with methylation levels,

HFD: rho = 0.829 , p < 0.05, R2 = 0.9635, CHD: rho = 1.000, p < 0.01, R2 = 0.9902.

a) % Global CpG methylation b) Global methylation vs. DNMT activity

57

Chapter 4

4 Discussion

The goal of this study was to examine the effects of perinatal HFD-exposure on the

transcriptional response to acute stressors in the brain during adulthood. Overall, we found

enhancements in anti-inflammatory signaling in response to CORT and/or LPS in both the

amygdala and hippocampus that appeared to be dependent on GR transcript levels. The diet-

dependence was the most apparent in the amygdala of CORT-challenged HFD males and

CORT/LPS-challenged HFD females, as we had previously found with non-stressed offspring

(Sasaki et al., 2013). Preliminary findings suggest impairments in mediators of DNA methylation

to be related to these diet-differences, since the DNMT activity of CORT-challenged HFD males

was significantly reduced, and global CpG methylation failed to decrease in response to CORT;

although the involvement of other means of epigenetic regulation, such as miRNAs or histone

modifications, cannot be discounted. Together, the findings suggest that early exposure to

maternal overnutrition may place offspring at particular risk for developing infections, and

behavioral/cognitive differences during stressful events encountered during adulthood,

depending on the nature of the stressor. We will start by discussing results from the CORT-

challenge.

4.1 Interpretation of results from CORT challenge

4.1.1 Diet x Sex differences in the Amygdala

Sorrells et al. (2009) suggest that GR is activated to suppress the immune response that initiates

in response to stress, which can be in the form of moderate, chronic doses of CORT, or acute

58

high doses of CORT (Dhabhar, 2000). However, CORT is also known to enhance the immune

response depending on the dose, duration, and type of tissue (Sorrells et al., 2009; Dhabhar,

2009; Bowers et al., 2008). For example, basal levels of CORT are necessary to induce immune

responses during a stressful event (Sorrells et al., 2009). This is comparable to how severe

injuries initially induce a systemic inflammatory response that is followed by an anti-

inflammatory response (Gouel-Chéron et al., 2012), where sepsis arises from simultaneous and

profound elevations of both responses (Hotchkiss et al., 2013; Boomer et al., 2011). GR is well-

established to elicit its immunosuppressive effects by initiating the transcription of anti-

inflammatory molecules including IkBa and MKP-1, and through preventing the transcription

factor NFkB from binding to its target gene binding-sites (Rhen and Cidlowski, 2005). This

includes the pro- and anti-inflammatory cytokines, IL-6 and IL-10 (Libermann and Baltimore,

1990; Saraiva and O'Garra, 2010).

An increased IL-6/IL-10 ratio was observed 3 hours post-CORT, supporting the literature

suggesting that CORT initially induces an immune response to stress. Furthermore, the

expression of GR, MKP-1, and IkBa were tightly correlated, in that perinatal HFD males

sustained higher expressions of each gene, while CHD males maintained relatively lower levels

of each. Analogous to hyperactive immune responses to injury, the simultaneous elevation of

pro- and anti-inflammatory transcripts in the brain may indicate an increased risk for mild-to-

high grades of septic encephalopathy, which is known to lead to long-term cognitive and

behavioral changes such as anxiety-like behavior, depression, and memory and learning deficits

(Calsavara et al., 2013; Ziaja, 2013).

Reduced negative feedback of the HPA-axis from the HFD males was also implicated by

the higher and increased transcription of CRH relative to CHD. This could be caused by the

59

elevations of IL-10 and GR in the amygdala relative to CHD, which are both known to activate

CRH in the hypothalamus and/or amygdala (Kolber et al., 2008; Smith et al., 1999). In the

amygdala, the activity of CRH has long been suggested to enhance anxiety (McGill et al., 2006;

Samaco et al., 2012), reinforcing the possibility of higher anxiety of HFD males responding to

CORT.

Comparing the IL-6/IL-10 ratio between sexes reveals that males sustain much greater

immunosuppressive activity in the absence of stress, and that they increase inflammation to

levels similar to females after being challenged with CORT [F(1,45) = 11.177, p < 0.01, Figure

16]. This supports common findings that females have basally active immune systems that are

more efficient at eliminating pathogens (Scotland et al., 2011; Rohleder et al., 2001). Hence,

during stress, females appear to have a higher threshold required to activate GR, as the levels of

IL-6 to IL-10 are already sufficient to respond to CORT, and does not need to further increase.

This would explain the lack of changes in transcript levels observed.

Figure 16. Comparison of IL-6/IL-10 ratios between CORT-challenged males and females in the

amygdala. Males sustain lower inflammatory activity, then reach levels equivalent to females

60

post-CORT. Bar **, *: p < 0.01, p < 0.05 post-hoc comparison within diet, ****: post-hoc

comparison within drug and diet.

On the other hand, HFD females have a significant increase in IL-6/IL-10 that co-occurs

with significant increases in IkBa, and decreased GR in response to CORT. It has been indicated

that the NFkB pathway can self-regulate by increasing the expression of IkBa independently of

GR (Sorrells et al., 2009). Therefore, the reduced transcription of GR may be inappropriately

causing elevations in IL-6/IL-10, while NFkB induces IkBa as a compensatory mechanism.

Under the assumption that transcript levels in the brain are representative of protein

levels in the plasma, the heightened IL-6/IL-10 ratio in responding to stress may be indicative of

depressive behavior according to a number of studies (Dhabhar et al., 2009; Fredericks et al.,

2009; Voorhees et al., 2013). Given that the periphery is known to send immune cells to the

blood-brain-barrier, cytokine receptors lining the vagus nerve and as recently discovered,

through lymphatic vessels connected to the brain (Louveau et al., 2015), to communicate and

propagate its inflammatory state, this could be a possibility (Maier and Watkins, 2012; Kronfol,

2003). Since MKP-1 is also a marker for depression, and it is elevated in HFD males, they may

also display more depressive behavior (Duric et al., 2010). Therefore, behavioral aberrations

potentially exist to a higher extent for both male and female HFD groups when responding to

stress. Behavioral studies will be needed to confirm this.

61

4.1.2 Females' distinct response in the Hippocampus

Here, we report very similar transcriptional responses between the amygdala and

hippocampus when responding to CORT, especially for males. On the other hand, CHD females

did not display any changes in transcript abundance in the amygdala, while transcription of many

genes reduced in response to CORT in the hippocampus. This appeared to relate to the CHD

group having higher initial transcript levels of these genes than the HFD group. While direct

comparisons of stress-invoked inflammatory changes has been sparse, the brain region-

specificity could be supported by a study that measured the activity and plasticity of subregions

of the hippocampus and amygdala in CHD rats following stressful exposures to a Morris Water

Maze test. It was discovered that long-term potentiation was differentially affected between the

two brain regions (Kavushansky et al., 2006). Since neuronal activity is regulated by cytokines,

the study may support the existence of distinct immune responses between the two regions

(Albensi and Mattson, 2000; Datson et al., 2013). Our results suggested that non-stressed HFD

females possess lower inflammatory activity in the hippocampus, but that they become more

active than CHD females in both limbic brain areas during stress with the higher levels of IkBa

and MKP-1, and exclusive enrichments of IL-6/IL-10 – again accompanied with lack of reduced

abundance of GR. However, considering that the levels become equivalent to those of CHD

females, it is arguable that GR is in fact meant to be functioning in a pro-inflammatory manner,

since CD11b and NFkB displayed reductions in transcript levels in a very similar manner to GR.

In either case, the transcript abundance of GR appears to coincide with differential responses

between the diet groups. The consequence this may have on performance in stressful learning,

memory, and fear-conditioning tasks such as the water maze may be interesting to examine.

Given that pro-inflammatory cytokines is induced and crucial for normal learning, and that

62

memory is impaired by excessively high levels (Williamson et al., 2011), HFD exposure would

likely have an effect in this cohort.

Taken together, perinatal HFD-exposed males in this study demonstrate exacerbated anti-

inflammatory responses, while HFD females show both higher pro- and anti-inflammatory

responses to CORT, which both co-occur with differences in transcription of GR. This could

suggest an increased susceptibility to infections and anxiety behavior for males following

physiological stress, and the risk for developing mild-to-severe grades of inflammatory-related

illnesses such as septic encephalopathy, which is characterized by anxiety, depression, and

cognitive impairments for both sexes.

4.2 Results from LPS challenge in the amygdala

Similar to the HFD females' response to CORT, both pro- and anti-inflammatory transcription

simultaneously increased for both HFD groups responding to LPS while the CHD groups

primarily increased in pro-inflammatory transcription. This again suggests an anti-inflammatory

bias on the part of the perinatal HFD-exposed animals, which occurs with the characteristic lack

of reductions in GR, as well as MR for males. The similarity to the CORT-challenged group is

likely due to the gradual secretion of CORT that naturally occurs after exposure to LPS for the

purpose of ameliorating the immune response - supporting the significance of GR in causing the

diet differences (Quan et al., 2000). Our finding contradicts two studies: One, that suggests MR

to have a pro-inflammatory effect under low-moderate CORT exposure (Changtong et al., 2012),

and another finding from Bilbo and Tsang (2010) where the protein expression of pro-

inflammatory cytokine, IL-1β were elevated in the hippocampus, and microglial activation was

enhanced both basally, and post-LPS challenge. In this study, we did not find significant effects

63

of diet in the hippocampus, nor did we find many in either the amygdala or hippocampus with

the dose used in that study (not shown). Going by CD11b levels, basal microglia concentrations

may have instead been reduced in the HFD-exposed males. Therefore, it's possible that the action

of MR may only be pro-inflammatory depending on the initial state of surrounding microglia.

Given the context-dependency of GR and MR (Sorrells et al., 2009), this could be the case, and

warrants further investigation. Put together, this would support the general idea that the

inflammatory, hence behavioral effects of perinatal HFD would depend on how it affects the

HPA-axis, as well the responsiveness of microglia separately, both of which could vary with

numerous factors such as sex, species strain, handling, or other variations in cohorts. Once more,

the coinciding elevation of pro- and anti-inflammatory transcript abundance may suggest

increased health risks with perinatal HFD-exposure, including grades of septic encephalopathy,

with the accompanying affect and cognitive impairments.

4.3 Results from CORT/LPS challenge in the amygdala

Although CORT is classically known to be immunosuppressive, there have been numerous

studies indicating that GR activation by acute or chronic stress can increase LPS-induced

expression of pro-inflammatory cytokines in limbic brain areas, excluding the hypothalamus

(Sorrells et al., 2009; Frank et al., 2009; Munhoz et al., 2006; de Pablos et al., 2006). This has

been shown to occur at least in part by GR-dependent proliferation and increased activation of

microglia, the immune cells of the brain (Nair and Bonneau, 2006; Frank et al., 2006), and by

lack of inductions of MKP-1 and/or IkBa (Munhoz et al., 2010). The change in function of GR is

speculated to occur because pro-inflammatory responses can be protective in neurons (e.g. when

facilitating hippocampal synaptic plasticity; Albensi and Mattson, 2000), while being damaging

to microglia (Sorrells et al., 2009). Since the pro-inflammatory effects usually occur when CORT

64

is given hours prior to LPS, and not simultaneously, we predicted that previous, perinatal HFD

exposure to CORT would be sufficient to provoke a similar response.

Compared to a high-dose LPS treatment alone (50 ug/kg), we indeed observed elevated

IL-6/IL-10 ratios with the addition of CORT for the CHD groups, with very high levels for the

CHD females. However, pro-inflammatory transcript levels were reduced for HFD females

relative to the CHD group, while it is less clear what is occurring for the HFD males, as IL-6/IL-

10 ratios are slightly higher for CHD males, but NFkB levels are higher for HFD males. In this

case, the CHD females maintain higher abundance of GR and MR than the HFD females -

further suggesting the involvement of GR in directing both pro- and anti-inflammatory signaling

depending on the stressor. The observation of GR decreasing, rather than being sustained,

supports past studies with LPS given after CORT - GR protein levels were shown to be reduced,

even though they were found to be necessary for increasing inflammation (Sorrells et al., 2009).

More support of the dependency on GR is the higher abundance of CD11b, a marker for

microglia, in the CORT/LPS-challenged CHD females relative to the HFD females. Further, the

increased CRH levels for CHD groups supports the enhanced LPS response as being a result of

lower negative feedback on the HPA-axis. Although the results suggest HFD females have an

elevated risk of developing infections against bacteria, their lower IL-6/IL-10 ratio and MKP-1

transcripts may suggest a decreased risk of sickness-induced depression – pointing to the delicate

balance that seems to exist between adaptive versus ‘maladaptive’ responses to stress, in that that

some levels of anxiety, for instance, might be necessary for learning, as previously implicated by

the maternal HFD study by Bilbo and Tsang (2010). Put together, perinatal HFD, combined with

CORT, impairs the pro-inflammatory, neuroprotective response to LPS in females.

65

4.4 Comparison of results with Sasaki et al. (2013) Maternal HFD Study

Here, we report that the majority of differences under baseline (non-injected) conditions occur in

the hippocampus of females, while none were apparent in the amygdala. The very opposite was

discovered in our laboratory’s previous study (Sasaki et al., 2013). Also, results between the

HFD and CHD males were similar in the hippocampus, but slightly different in the amygdala. In

this study, only MR and CD11b were different under baseline (HFD < CHD), whereas some

differences in GR, pro-, and anti-inflammatory transcript levels had been shown before. While

the same diet paradigms, primer sets, and storage/extraction procedures were used, we could not

account for any differences in handling, time of sacrifice, or exposures to cues from challenged

rodents within the room, which may have all contributed to these differing results. In summary,

Figure 17 re-illustrates the CORT/LPS pathway with respect to each gene analyzed in this study

with coloured arrows to indicate the effects of perinatal HFD-exposure we observed.

66

Figure 17. Simplified diagram of the effect of Perinatal HFD on the general CORT/LPS

pathway. Overview: An Inflammatory-challenge such as LPS activates NFkB, which

translocates to the nucleus to induce the transcription of pro-inflammatory cytokines such as IL-6

to initiate the immune response (Libermann and Baltimore, 1990). Over time (dashed arrows),

CORT is secreted to activate GR and repress NFkB activity by blocking its binding to consensus

sequences, and through inducing the expression of anti-inflammatory molecules such as IkBa

and MKP-1 (Rhen and Cidlowski, 2005). NFkB also negatively inhibits itself by progressively

increasing the expression of anti-inflammatory molecules (Sorrells et al., 2009). In the brain,

prior CORT exposure has been shown to enhance immune responses to LPS (Frank et al., 2006).

67

The green arrows indicate where we predominantly observed enhancements in activity as a result

of perinatal HFD-exposure (immunosuppression by CORT), while the orange arrow indicates

where it is reduced (pro-inflammation by CORT). GR is bolded to indicate its central role in the

diet-differences.

4.5 Maternal HFD transmission of CORT as possible mechanism for differential GC

sensitivity

In order to explain these effects, it would be reasonable to question whether perinatal exposure to

HFD may be priming offspring to respond to CORT later in life by directly increasing fetal

exposure to maternal circulating levels of CORT. Indeed, maternal HFD has been reported to

increase plasma CORT and decrease 11β-HSD-2 expression and activity in rodents, which is an

enzyme that catalyzes the inactivation of maternal GCs before they are transferred to the fetus

(Bellisario et al., 2015). Moreover, these conditions are known to lead to reduced gestational

body weight (Bellisario et al., 2015; Lindsay et al., 1996), which independently results in much

of the downstream mental health outcomes in humans (Grissom and Reyes, 2013; Rivera et al.,

2015). The HFD-exposed offspring showed reduced body weight at birth, suggesting that a

heightened exposure to CORT may have altered the way the offspring respond to CORT and

CORT/LPS during adulthood - with receptors of males being able to bind a higher concentration

of CORT, while females reduce transcript levels more intensely with the addition of LPS.

Potential evidence for this is the reduction of basal transcript levels of MR that we

observed in the amygdala of males (Table 6), which should theoretically increase the occupancy

of CORT to GR. This may have permanently conditioned GR to bind higher concentrations of

CORT before downregulating itself during a stress response. Alternatively, the early exposures

68

to GCs could have changed the expression of corticosteroid-binding globulin (CBG), which

escorts CORT through the blood and keeps it in an inactive state (Dhillo et al., 2002). If CBG

levels are higher in HFD-exposed animals, then the downregulation of GR may occur at later

time-points due to decreased contact with CORT - thereby prolonging HPA-axis activity.

However, this is less likely, as we saw higher pro- and anti-inflammatory activity, which

suggests that CORT is activating GR more frequently than with CHD animals. Nonetheless, we

cannot discount the possibility that the microglia of HFD-exposed animals had become more

sensitive to GR, potentially through impaired MeCP2 functionality, as we will discuss below.

Overall, direct perinatal overexposure to CORT offers one possible mechanism for the diet-

dependent transcriptional responses to CORT and CORT/LPS observed in this study.

4.6 Role of Epigenetic Regulators: MeCP2 as potential hub for manifestation of diet- and

sex- differences

In numerous, but not all instances, the transcript changes of epigenetic regulators resemble, or

‘cluster’ with that of GR and its downstream target genes. This can especially be observed for

CORT-challenged males, and CORT/LPS-challenged females in the amygdala. In both cases,

DNMT1 levels were commonly clustered with those of the genes - very similar to a study

between anxious humans which showed concurrent increases in gene expression of all DNMT

isoforms and IL-6 in peripheral blood mononuclear cells, with self-reports of anxiety (Murphy et

al., 2014). This observation provoked us to examine the DNMT activity of these cohorts, and it

was found that HFD males displayed greatly reduced total DNMT activity that tightly negatively

correlated with DNMT1 transcript levels and global CpG methylation levels. However, DNMT1

protein levels were not different across diet and treatment groups, and did not correlate to either

activity or transcript levels. This more closely mirrors what has been found in previous studies of

69

DNMT activity in the amygdala and medial preoptic area of the hypothalamus, where levels of

protein did not change in the presence of differing DNMT activity, suggesting alternate means of

changing activity, such as phosphorylation and/or reduced binding affinity to interacting partners

(McCarthy et al., 2010; Nugent et al., 2015). Reduced interactions with MeCP2 due to changes

in its phosphorylation state would be a strong candidate for explaining the lowered activity, as

this was observed to occur for maternal HFD-exposed groups that showed impaired DNMT

activity in the prefrontal cortex in another study (McKee et al. 2014). Hence, the changes in

transcription of DNMT1 may merely be capturing the effects on GR without affecting actual

expression or function, as GR has been found to target the DNMTs downstream when

responding to CORT (Urb et al., 2015). Co-immunoprecipitation (Co-IP) studies will be needed

to clarify the effect of reduced DNMT activity on interactions with MeCP2 and GR.

On the other hand, there were no differences in DNMT activity across diet and drug for

the CORT/LPS-injected females. We speculated that perhaps, then, demethylase activity may be

responsible for the diet-differences observed in responding to CORT/LPS. This was a possibility,

considering how both DNMTs and the demethylase enzyme, Tet (specifically, Tet1) require

interactions with MeCP2 for its activity (Kimura and Shiota, 2003; Cartron et al., 2013), and

MeCP2 itself represses genes in the microglia induced by GCs and TNF-α, another pro-

inflammatory stimulus (Cronk et al., 2015). There are also findings that Gadd45b modulates the

expression of pro-inflammatory cytokines such as IL-6 in the amygdala (Kigar et al., 2015), and

that adult females, but not males show decreases in Gadd45b expression in response to early

caregiver maltreatment (Blaze and Roth, 2013) – which we have also observed in the amygdala

in response to perinatal HFD. Hence, future studies on measurements of demethylase activity

and Co-IP will be required to confirm this hypothesis. It is particularly apparent that more focus

70

needs to be placed on MeCP2 with studies involving effects of perinatal stress on future stress

challenges.

4.7 Explaining the effects of DNMT activity on Global CpG methylation levels

Since the DNMT activity kits used specifically measured the ability of DNMTs to methylate a

DNA substrate, it was initially surprising to observe the decreased activity occurring with no

changes in global methylation levels for the CORT-challenged HFD males, whereas the levels

significantly reduced for the CHD males. To begin with, reductions in methylation can only be

occurring through active demethylase activity, or passively, through replication events where

DNMT1 does not copy the methylation marks onto newly synthesized DNA. Since DNMT

activity was not reduced for the CHD males, the latter does not appear to apply, and the results

may reinforce the existence of impairments in both methylase and demethylase activity through

perinatal HFD-exposure, where GR-dependent effects on inflammatory pathways depend on the

type of stressor and sex of the stressed individual. In which case, DNMT activity seems essential

for responding to CORT in males, important enough to incur interindividual differences in global

CpG methylation levels, but not for CORT/LPS-challenged females. In order to clarify that the

absence or presence of DNA methylation is leading to the diet-dependent transcriptional

responses to each challenge, future bisulfite pyrosequencing studies will have to be conducted

for the genes studied, particularly GR, which uses epigenetic modifications to down-regulate

itself after activation with CORT (Ramamoorthy and Cidlowski, 2013). To attain a greater

picture of our findings, microarray studies should also be performed to examine potential

discrepancies between transcript responses, and actual gene expression changes, since mRNA

stability can be compromised by miRNA, another epigenetic mechanism.

71

4.8 Conclusion

To summarize, we report that rats exposed to perinatal HFD respond to the acute stress of

CORT, LPS, and CORT/LPS with predominantly enhanced immunosuppressive activity seeming

to relate to GR transcript abundance, and altered abundance of genes associated with anxiety and

depression in a sex and brain region-specific manner. As GR is known to interact with epigenetic

mediators to regulate the transcription of target genes when responding to stress, including its

own downregulation, impaired DNA methylase and demethylase activity through reduced

associations with MeCP2 may at least in part explain the differential responses. Future

experiments including Co-IP, bisulfite pyrosequencing, and more protein activity and

quantification assays will be required to confirm this hypothesis.

References

Afari, N., & Buchwald, D. (2003). Chronic fatigue syndrome: A review. American Journal of

Psychiatry, 160(2), 221–236. doi:10.1176/appi.ajp.160.2.221

Albensi, B. C., & Mattson, M. P. (2000). Evidence for the involvement of TNF and NF-kappaB

in hippocampal synaptic plasticity. Synapse (New York, N.Y.), 35(2), 151–159.

doi:10.1002/(SICI)1098-2396(200002)35:2<151::AID-SYN8>3.0.CO;2-P

Anacker, C., Zunszain, P. a, Carvalho, L. a, & Pariante, C. M. (2012). Europe PMC Funders

Group The glucocorticoid receptor : Pivot of depression and of antidepressant treatment ?,

36(3), 415–425. doi:10.1016/j.psyneuen.2010.03.007.The

72

Bake, S., Selvamani, A., Cherry, J., & Sohrabji, F. (2014). Blood brain barrier and

neuroinflammation are critical targets of IGF-1-mediated neuroprotection in stroke for

middle-aged female rats. PLoS ONE, 9(3). doi:10.1371/journal.pone.0091427

Barker, E. D., Kirkham, N., Ng, J., & Jensen, S. K. G. (2013). Prenatal maternal depression

symptoms and nutrition, and child cognitive function. The British Journal of Psychiatry :

The Journal of Mental Science, 203(6), 417–21. doi:10.1192/bjp.bp.113.129486

Bartha, J. L., Romero-Carmona, R., & Comino-Delgado, R. (2003). Inflammatory cytokines in

intrauterine growth retardation. Acta Obstetricia et Gynecologica Scandinavica, 82(12),

1099–1102. doi:10.1046/j.1600-0412.2003.00259.x

Bellisario, V., Panetta, P., Balsevich, G., Baumann, V., Noble, J., Raggi, C., … Cirulli, F.

(2015). Maternal high-fat diet acts as a stressor increasing maternal glucocorticoids’

signaling to the fetus and disrupting maternal behavior and brain activation in C57BL/6J

mice. Psychoneuroendocrinology, 60, 138–150. doi:10.1016/j.psyneuen.2015.06.012

Benros, M. E., Nielsen, P. R., Nordentoft, M., Eaton, W. W., Dalton, S. O., & Mortensen, P. B.

(2011). Autoimmune diseases and severe infections as risk factors for schizophrenia: A

30-year population-based register study. American Journal of Psychiatry, 168(12), 1303–

1310. doi:10.1176/appi.ajp.2011.11030516

Bilbo, S. D., & Schwarz, J. M. (2009). Early-life programming of later-life brain and behavior: a

critical role for the immune system. Frontiers in Behavioral Neuroscience, 3(August), 1–

14. doi:10.3389/neuro.08.014.2009

73

Bilbo, S. D., & Tsang, V. (2010). Enduring consequences of maternal obesity for brain

inflammation and behavior of offspring. The FASEB Journal : Official Publication of the

Federation of American Societies for Experimental Biology, 24(6), 2104–2115.

doi:10.1096/fj.09-144014

Bilbo, S. D., Yirmiya, R., Amat, J., Paul, E. D., Watkins, L. R., & Maier, S. F. (2008). Bacterial

infection early in life protects against stressor-induced depressive-like symptoms in adult

rats. Psychoneuroendocrinology, 33(3), 261–9. doi:10.1016/j.psyneuen.2007.11.008

Binder, E. B. (2009). The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the

pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology,

34, S186–S195. doi:10.1016/j.psyneuen.2009.05.021

Blaze, J., & Roth, T. L. (2013). Exposure to caregiver maltreatment alters expression levels of

epigenetic regulators in the medial prefrontal cortex. International Journal of

Developmental Neuroscience, 31(8). doi:10.1016/j.ijdevneu.2013.10.001

Bolton, J. L., & Bilbo, S. D. (2014). Developmental programming of brain and behavior by

perinatal diet: focus on inflammatory mechanisms. Dialogues in Clinical Neuroscience,

16(3), 307–20. Retrieved from

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4214174&tool=pmcentrez&r

endertype=abstract

Boomer, J. S. (2011). Immunosuppression in Patients Who Die of Sepsis and Multiple Organ

Failure. JAMA: The Journal of the American Medical Association, 306(23), 2594.

doi:10.1001/jama.2011.1829

74

Bourre, J. M., Gozlan-Devillierre, N., Morand, O., & Baumann, N. (1979). Importance of

exogenous saturated fatty acids during brain development and myelination in mice.

Annales de Biologie Animale Biochimie Biophysique, 19(1B), 173–180.

doi:10.1051/rnd:19790205

Bowers, S. L., Bilbo, S. D., Dhabhar, F.S., & Nelson, R.J. (2008) Stressor-specific alterations in

corticosterone and immune responses in mice. Brain, Behavior, and Immunity, 22, 105–

113. doi:10.1016/j.bbi.2007.07.012

Calsavara, A. C., Rodrigues, D. H., Miranda, A. S., Costa, P. a., Lima, C. X., Vilela, M. C., …

Teixeira, A. L. (2013). Late anxiety-like behavior and neuroinflammation in mice

subjected to sublethal polymicrobial sepsis. Neurotoxicity Research, 24(2), 103–108.

doi:10.1007/s12640-012-9364-1

Can,O. D., Ulupinar, E.,Ozkay, U. D., Yegin, B., andOzturk, Y. (2012). The effect of simvastatin

treatment on behavioral parameters, cognitive performance, and hippocampal

morphology in rats fed a standard or a high-fat diet. Behav. Pharmacol. 23, 582–592. doi:

10.1097/FBP.0b013e328356c3f2

Canani, R. B., Costanzo, M. Di, Leone, L., Bedogni, G., Brambilla, P., Cianfarani, S., …

Agostoni, C. (2011). Epigenetic mechanisms elicited by nutrition in early life Nutrition

Research Reviews Nutrition Research Reviews, 198–205.

doi:10.1017/S0954422411000102

Cartron, P.-F., Nadaradjane, A., Lepape, F., Lalier, L., Gardie, B., & Vallette, F. M. (2013).

Identification of TET1 Partners That Control Its DNA-Demethylating Function. Genes &

Cancer, 4(5-6), 235–41. doi:10.1177/1947601913489020

75

Chantong, B., Kratschmar, D. V, Nashev, L. G., Balazs, Z., & Odermatt, A. (2012).

Mineralocorticoid and glucocorticoid receptors differentially regulate NF-kappaB activity

and pro-inflammatory cytokine production in murine BV-2 microglial cells. Journal of

Neuroinflammation, 9(1), 260. doi:10.1186/1742-2094-9-260

Chiba, S., Numakawa, T., Ninomiya, M., Richards, M. C., Wakabayashi, C., & Kunugi, H.

(2012). Chronic restraint stress causes anxiety- and depression-like behaviors,

downregulates glucocorticoid receptor expression, and attenuates glutamate release

induced by brain-derived neurotrophic factor in the prefrontal cortex. Progress in Neuro-

Psychopharmacology and Biological Psychiatry, 39(1), 112–119.

doi:10.1016/j.pnpbp.2012.05.018

Chmielarz, P., Kuśmierczyk, J., Parlato, R., Schütz, G., Nalepa, I., & Kreiner, G. (2013).

Inactivation of glucocorticoid receptor in noradrenergic system influences anxiety- and

depressive-like behavior in mice. PloS One, 8(8), e72632.

doi:10.1371/journal.pone.0072632

Colman, I., Jones, P. B., Kuh, D., Weeks, M., Naicker, K., Richards, M., & Croudace, T. J.

(2014). Early development, stress and depression across the life course: pathways to

depression in a national British birth cohort. Psychological Medicine, 44, 2845–2854.

doi:10.1017/S0033291714000385

Cronk, J. C., Litvak, V., Kipnis, J., Derecki, C., Ji, E., Xu, Y., … Baker, W. (2015). Methyl-CpG

Binding Protein 2 Regulates Microglia and Macrophage Gene Expression in Response to

Article Methyl-CpG Binding Protein 2 Regulates Microglia and Macrophage Gene

Expression in Response to Inflammatory Stimuli, 679–691.

76

Crudo, A., Petropoulos, S., Moisiadis, V. G., Iqbal, M., Kostaki, A., Machnes, Z., … Matthews,

S. G. (2012). Prenatal synthetic glucocorticoid treatment changes DNA methylation states

in male organ systems: Multigenerational effects. Endocrinology, 153(7), 3269–3283.

doi:10.1210/en.2011-2160

Datson, N. A., Van Den Oever, J. M. E., Korobko, O. B., Magarinos, A. M., De Kloet, E. R., &

McEwen, B. S. (2013). Previous history of chronic stress changes the transcriptional

response to glucocorticoid challenge in the dentate gyrus region of the male rat

hippocampus. Endocrinology, 154(C), 3261–3272. doi:10.1210/en.2012-2233

de Pablos, R.M., Villaran, R.F., Arguelles, S., Herrera, A.J., Venero, J.L., Ayala, A., Cano, J., &

Machado, A. (2006). Stress increases vulnerability to inflammation in the rat prefrontal

cortex. Journal of Neuroscience, 26(21), 5709–5719. doi:10.1523/JNEUROSCI.0802-

06.2006

de Souza, C. T., Araujo, E.P., Bordin, S., Ashimine, R., Zollner, R. L., Boschero, A. C., Saad, M.

J., & Velloso, L.A. (2005) Consumption of a fat-rich diet activates a proinflammatory

response and induces insulin resistance in the hypothalamus. Endocrinology, 146, 4192–

4199. doi:10.1210/en.2004-1520

Dhabhar, F. S. (2000). Acute Stress Enhances While Chronic Stress Suppresses Skin Immunity:

The Role of Stress Hormones and Leukocyte Trafficking. Annals of the New York

Academy of Sciences, 917, 876–893. doi: 10.1111/j.1749-6632.2000.tb05454.x

Dhabhar, F. S. (2009). Enhancing versus suppressive effects of stress on immune function:

Implications for immunoprotection and immunopathology. Neuroimmunomodulation, 16,

300–317. doi:10.1159/000216188

77

Dhabhar, F. S., Burke, H. M., Epel, E. S., Mellon, S. H., Rosser, R., Reus, V. I., & Wolkowitz,

O. M. (2009). Low serum IL-10 concentrations and loss of regulatory association

between IL-6 and IL-10 in adults with major depression. Journal of Psychiatric

Research, 43(11), 962–969. doi:10.1016/j.jpsychires.2009.05.010

Dhillo, W. S., Kong, W. M., Le Roux, C. W., Alaghband-Zadeh, J., Jones, J., Carter, G., …

O’Shea, D. (2002). Cortisol-binding globulin is important in the interpretation of

dynamic tests of the hypothalamic-pituitary-adrenal axis. European Journal of

Endocrinology, 146, 231–235. doi:10.1530/eje.0.1460231

Djordjevic, J., Djordjevic, a., Adzic, M., Niciforovic, a., & Radojcic, M. B. (2010). Chronic

stress differentially affects antioxidant enzymes and modifies the acute stress response in

liver of wistar rats. Physiological Research, 59(11), 729–736.

Doherty, a S., Mann, M. R., Tremblay, K. D., Bartolomei, M. S., & Schultz, R. M. (2000).

Differential effects of culture on imprinted H19 expression in the preimplantation mouse

embryo. Biology of Reproduction, 62(6), 1526–1535. doi:10.1095/biolreprod62.6.1526

Driessler, F., Venstrom, K., Sabat, R., Asadullah, K., & Schottelius, a J. (2004). Molecular

mechanisms of interleukin-10-mediated inhibition of NF-kappaB activity: a role for p50.

Clinical and Experimental Immunology, 135(1), 64–73. doi:10.1046/j.1365-

2249.2004.02342.x

Duan, S. Z., Zhang, W. C., & Zheng, X. J. (2013). An Update on Actions of Mineralocorticoid

Receptor in the Cardiovascular System : From Clinical Studies to Basic Research, 6(2),

91–94. doi:10.7156/najms.2013.0602091

78

Dunn, G.A., & Bale, T.L. (2009) Maternal high-fat diet promotes body length increases and

insulin insensitivity in second-generation mice. Endocrinology 150(11), 4999–5009. doi:

10.1210/en.2009-0500

Durairaj, R. V., & Koilmani, E. R. (2014). Environmental enrichment modulates glucocorticoid

receptor expression and reduces anxiety in Indian field male mouse Mus booduga through

up-regulation of microRNA-124a. General and Comparative Endocrinology, 199, 26–32.

doi:10.1016/j.ygcen.2014.01.005

Duric, V., Banasr, M., Licznerski, P., Schmidt, H. D., Stockmeier, C. a, Simen, A. a, … Duman,

R. S. (2010). A negative regulator of MAP kinase causes depressive behavior. Nature

Medicine, 16(11), 1328–1332. doi:10.1038/nm.2219

El-Haschimi, K., Pierroz, D. D., Hileman, S. M., Bjorbaek, C., Flier, J. S. (2000). Two defects

contribute to hypothalamic leptin resistance in mice with diet-induced obesity. Journal of

Clinical Investigation, 105, 1827–1832. doi:10.1172/JCI9842

Fagiolini, M., Jensen, C. L., & Champagne, F. a. (2009). Epigenetic influences on brain

development and plasticity. Current Opinion in Neurobiology, 19(2), 207–12.

doi:10.1016/j.conb.2009.05.009

Fagerås, M., Jenmalm, M. C., Garofalo, P., Böttcher, M. F., Garofalo, R. P., & Björkstén,

B. (1999). Cytokines in breast milk from allergic and nonallergic mothers. Pediatric

Research, 47(1), 157–162. doi:10.1203/00006450-200005000-00006

79

Fatemi, M., Hermann, A., Gowher, H., & Jeltsch, A. (2002). Dnmt3a and Dnmt1 functionally

cooperate during de novo methylation of DNA. European Journal of Biochemistry,

269(20), 4981–4984. doi:10.1046/j.1432-1033.2002.03198.x

Feldker, D. E. M., Morsink, M. C., Veenema, A. H., Datson, N. A., Proutski, V., Lathouwers, D.,

… Vreugdenhil, E. (2006). The effect of chronic exposure to highly aggressive mice on

hippocampal gene expression of non-aggressive subordinates. Brain Research, 1089(1),

10–20. doi:10.1016/j.brainres.2006.02.110

Flegal, K. M., Carroll, M. D., Ogden, C. L., & Curtin, L. R. (2010). Prevalence and Trends in

Obesity Among US Adults, 1999-2008. JAMA, 303(3), 235–41. doi:

10.1001/jama.2009.2014.

Frank, M. G., Baratta, M. V., Sprunger, D. B., Watkins, L. R., & Maier, S. F. (2006). Microglia

serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-

inflammatory cytokine responses. Brain, Behavior, and Immunity, 21(1), 47–59.

doi:10.1016/j.bbi.2006.03.005

Frank, M.G., Miguel, Z.D., Watkins, L.R., & Maier, S.F. (2009). Prior exposure to

glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses

to E. coli lipopolysaccharide. Brain, Behavior, and Immunity, 24(1), 19–30. doi:

10.1016/j.bbi.2009.07.008.

Fredericks, C. a., Drabant, E. M., Edge, M. D., Tillie, J. M., Hallmayer, J., Ramel, W., …

Dhabhar, F. S. (2010). Healthy young women with serotonin transporter SS

polymorphism show a pro-inflammatory bias under resting and stress conditions. Brain,

Behavior, and Immunity, 24(3), 350–357. doi:10.1016/j.bbi.2009.10.014

80

Freeman, D. J. (2010). Effects of maternal obesity on fetal growth and body composition:

implications for programming and future health. Seminars in Fetal and Neonatal

Medicine, 15(2), 113–118. doi:10.1016/j.siny.2009.09.001

Giriko, C. A., Andreoli, C. A., Mennitti, L. V., Hosoume, L. F., Souto Tdos, S., Silva, A. V., et

al. (2013). Delayed physical and neurobehavioral development and increased aggressive

and depression-like behaviors in the rat offspring of dams fed a high-fat diet.

International Journal of Developmental Neuroscience, 31, 731–739. doi:

10.1016/j.ijdevneu.2013.09.001

Godfrey, K. M., Gluckman, P. D., & Hanson, M. A. (2010). Developmental origins of metabolic

disease: life course and intergenerational perspectives. Trends in endocrinology and

metabolism, 21(4), 199-205. doi: 10.1016/j.tem.2009.12.008.

Gouel-Chéron, A., Allaouchiche, B., Guignant, C., Davin, F., Floccard, B., & Monneret, G.

(2012). Early interleukin-6 and slope of monocyte human leukocyte antigen-DR: A

powerful association to predict the development of sepsis after major trauma. PLoS ONE,

7(3). doi:10.1371/journal.pone.0033095

Grissom, N. M., George, R., & Reyes, T. M. (2015). The hypothalamic transcriptional response

to stress is severely impaired in offspring exposed to adverse nutrition during gestation.

Neuroscience, (July). doi:10.1016/j.neuroscience.2015.07.022

Grissom, N. M., & Reyes, T. M. (2013). Gestational overgrowth and undergrowth affect

neurodevelopment: Similarities and differences from behavior to epigenetics.

International Journal of Developmental Neuroscience, 31(6), 406–414.

doi:10.1016/j.ijdevneu.2012.11.006

81

Heim, C., Newport, D. J., Heit, S., Graham, Y. P., Wilcox, M., Bonsall, R., … Nemeroff, C. B.

(2000). Pituitary-adrenal and autonomic responses to stress in women after sexual and

physical abuse in childhood. JAMA : The Journal of the American Medical Association,

284(5), 592–597. doi:joc92111 [pii]

Herrero, A. I., Sandi, C., & Venero, C. (2006). Individual differences in anxiety trait are related

to spatial learning abilities and hippocampal expression of mineralocorticoid receptors.

Neurobiology of Learning and Memory, 86(2), 150–9. doi:10.1016/j.nlm.2006.02.001

Hotchkiss, R. S., Monneret, G., & Payen, D. (2013). Sepsis-induced immunosuppression: from

cellular dysfunctions to immunotherapy. Nature Reviews. Immunology, 13(12), 862–74.

doi:10.1038/nri3552

Hsieh, C.-L. (2005). The de novo methylation activity of Dnmt3a is distinctly different than that

of Dnmt1. BMC Biochemistry, 6, 6. doi:10.1186/1471-2091-6-6

Hyman, S. E. (2009). How adversity gets under the skin. Nature Neuroscience, 12(3), 241–243.

doi:10.1038/nn0309-241

Jeltsch, A., & Jurkowska, R. Z. (2014). New concepts in DNA methylation. Trends in

Biochemical Sciences, 39(7), 310–318. doi:10.1016/j.tibs.2014.05.002

Jones, P. A, & Takai, D. (2001). The role of DNA methylation in mammalian epigenetics.

Science (New York, N.Y.), 293(5532), 1068–1070. doi:10.1126/science.1063852

Jones, P. A. (2012). Functions of DNA methylation: islands, start sites, gene bodies and beyond.

Nature Reviews. Genetics, 13(7), 484–92. doi:10.1038/nrg3230

82

Juruena, M. F., Pariante, C. M., Papadopoulos, A. S., Poon, L., Lightman, S., & Cleare, A. J.

(2013). The role of mineralocorticoid receptor function in treatment-resistant depression.

Journal of Psychopharmacology (Oxford, England), 27(12), 1169–79.

doi:10.1177/0269881113499205

Kanatsou, S., Kuil, L. E., Arp, M., Oitzl, M. S., Harris, A. P., Seckl, J. R., … Joels, M. (2015).

Overexpression of mineralocorticoid receptors does not affect memory and anxiety-like

behavior in female mice Article type : Received on : Accepted on : Frontiers website

link : Citation : Overexpression of mineralocorticoid receptors does not affect memo.

Frontiers in Behavioral Neuroscience, 9(July), 1–9. doi:10.3389/fnbeh.2015.00182

Kang, S. S., Kurti, A., Fair, D. a, & Fryer, J. D. (2014). Dietary intervention rescues maternal

obesity induced behavior deficits and neuroinflammation in offspring. Journal of

Neuroinflammation, 11(1), 156. doi:10.1186/s12974-014-0156-9

Karimi, M., Johansson, S., & Ekström, T. J. (2006). Using LUMA: A luminometric-based assay

for global DNA-methylation. Epigenetics, 1(1), 45–48. doi:10.4161/epi.1.1.2587

Kavushansky, A., Vouimba, R. M., Cohen, H., & Richter-Levin, G. (2006). Activity and

plasticity in the CA1, the dentate gyrus, and the amygdala following controllable vs.

uncontrollable water stress. Hippocampus, 16(1), 35–42. doi:10.1002/hipo.20130

Kigar, S. L., Chang, L., & Auger, A. P. (2015). Gadd45b is an epigenetic regulator of juvenile

social behavior and alters local pro-inflammatory cytokine production in the rodent

amygdala. Brain, Behavior, and Immunity, 46, 60–9. doi:10.1016/j.bbi.2015.02.018

83

Kimura, H., & Shiota, K. (2003). Methyl-CpG-binding protein, MeCP2, is a target molecule for

maintenance DNA methyltransferase, Dnmt1. Journal of Biological Chemistry, 278(7),

4806–4812. doi:10.1074/jbc.M209923200

Klok, M. D., Giltay, E. J., Van der Does, a J. W., Geleijnse, J. M., Antypa, N., Penninx, B. W. J.

H., … DeRijk, R. H. (2011). A common and functional mineralocorticoid receptor

haplotype enhances optimism and protects against depression in females. Translational

Psychiatry, 1(12), e62. doi:10.1038/tp.2011.59

Kohli, R. M., & Zhang, Y. (2013). TET enzymes, TDG and the dynamics of DNA

demethylation. Nature, 502(7472), 472–9. doi:10.1038/nature12750

Kolber, B. J., Roberts, M. S., Howell, M. P., Wozniak, D. F., Sands, M. S., & Muglia, L. J.

(2008). Central amygdala glucocorticoid receptor action promotes fear-associated CRH

activation and conditioning. Proceedings of the National Academy of Sciences of the

United States of America, 105(33), 12004–12009. doi:10.1073/pnas.0803216105

Koldzic-Zivanovic, N., Tu, H., Juelich, T. L., Rady, P. L., Tyring, S. K., Hudnall, S. D., …

Hughes, T. K. (2006). Regulation of adrenal glucocorticoid synthesis by interleukin-10:

A preponderance of IL-10 receptor in the adrenal zona fasciculata. Brain, Behavior, and

Immunity, 20(5), 460–468. doi:10.1016/j.bbi.2005.09.003

Kolodkin, M. H., & Auger, A. P. (2011). Sex difference in the expression of DNA

methyltransferase 3a (DNMT3a) in the rat amygdala during development. Journal of

Neuroendocrinology, 23(7), 577–583. doi:10.1111/j.1365-2826.2011.02147.x

84

Korte, S. M. (2001). Corticosteroids in relation to fear, anxiety and psychopathology.

Neuroscience and Biobehavioral Reviews, 25(2), 117–142. doi:10.1016/S0149-

7634(01)00002-1

Korte, S. M., Korte-Bouws, G. a, Koob, G. F., De Kloet, E. R., Bohus, B., Neurosciences, C., …

Laboratories, S. (1996). Mineralocorticoid and glucocorticoid receptor antagonists in

animal models of anxiety. Pharmacology, Biochemistry, and Behavior, 54(1), 261–7.

doi:0091-3057(95)02172-8 [pii]

Kossintseva, I., Wong, S., Johnstone, E., Guilbert, L., Olson, D. M., & Mitchell, B. F. (2006).

Proinflammatory cytokines inhibit human placental 11beta-hydroxysteroid

dehydrogenase type 2 activity through Ca2+ and cAMP pathways. American Journal of

Physiology. Endocrinology and Metabolism, 290(2), E282–8.

doi:10.1152/ajpendo.00328.2005

Krakowiak, P., Walker, C. K., Bremer, a. a., Baker, a. S., Ozonoff, S., Hansen, R. L., & Hertz-

Picciotto, I. (2012). Maternal Metabolic Conditions and Risk for Autism and Other

Neurodevelopmental Disorders. Pediatrics, 129(5), e1121–e1128.

doi:10.1542/peds.2011-2583

Kronfol, Z. [Ed]. (2003). Cytokines and mental health. Cytokines and Mental Health. Kluwer

Academic Publishers. Retrieved from:

http://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=psyc4&NEWS=N&AN

=2003-00899-000.

Ladd, C. O., Huot, R. L., Thrivikraman, K. V, Nemeroff, C. B., & Plotsky, P. M. (2004). Long-

term adaptations in glucocorticoid receptor and mineralocorticoid receptor mRNA and

85

negative feedback on the hypothalamo-pituitary-adrenal axis following neonatal maternal

separation. Biological Psychiatry, 55(4), 367–75. doi:10.1016/j.biopsych.2003.10.007

Lai, M., Horsburgh, K., Bae, S.-E., Carter, R. N., Stenvers, D. J., Fowler, J. H., … Macleod, M.

R. (2007). Forebrain mineralocorticoid receptor overexpression enhances memory,

reduces anxiety and attenuates neuronal loss in cerebral ischaemia. The European Journal

of Neuroscience, 25(6), 1832–42. doi:10.1111/j.1460-9568.2007.05427.x

Levitt, N. S., Lindsay, R. S., Holmes, M. C., & Seckl, J. R. (1996). Dexamethasone in the last

week of pregnancy attenuates hippocampal glucocorticoid receptor gene expression and

elevates blood pressure in the adult offspring in the rat. Neuroendocrinology, 64(6), 412-

8. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8990073

Li, M., Han, F., & Shi, Y. (2011). Expression of locus coeruleus mineralocorticoid receptor and

glucocorticoid receptor in rats under single-prolonged stress. Neurological Sciences,

32(4), 625–631. doi:10.1007/s10072-011-0597-1

Libermann, T. a, & Baltimore, D. (1990). Activation of interleukin-6 gene expression through

the NF-kappa B transcription factor. Molecular and Cellular Biology, 10(5), 2327–2334.

doi:10.1128/MCB.10.5.2327.Updated

Lillycrop, K. a, Phillips, E. S., Torrens, C., Hanson, M. a, Jackson, A. a, & Burdge, G. C. (2008).

Feeding pregnant rats a protein-restricted diet persistently alters the methylation of

specific cytosines in the hepatic PPAR alpha promoter of the offspring. The British

Journal of Nutrition, 100(2), 278–282. doi:10.1017/S0007114507894438

86

Lillycrop, K. a, Slater-Jefferies, J. L., Hanson, M. a, Godfrey, K. M., Jackson, A. a, & Burdge,

G. C. (2007). Induction of altered epigenetic regulation of the hepatic glucocorticoid

receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests

that reduced DNA methyltransferase-1 expression is involved in impaired DNA

methylation and . The British Journal of Nutrition, 97(6), 1064–73.

doi:10.1017/S000711450769196X

Louveau, A., Smirnov, I., Keyes, T. J., Eccles, J. D., Rouhani, S. J., Peske, J. D., … Kipnis, J.

(2015). Structural and functional features of central nervous system lymphatic vessels.

Nature. doi:10.1038/nature14432

MacKenzie, E. M., Odontiadis, J., Le Mellédo, J. M., Prior, T. I., & Baker, G. B. (2007). The

relevance of neuroactive steroids in schizophrenia, depression, and anxiety disorders.

Cellular and Molecular Neurobiology, 27(5), 541–574. doi:10.1007/s10571-006-9086-0

Maier, S. F., & Watkins, L. R. (2012). Consequences of the Inflamed Brain. Retrieved from The

Dana Foundation website:

http://www.dana.org/Publications/ReportOnProgress/Consequences_of_the_Inflamed_Br

ain/

Makino, S., Hashimoto, K., & Gold, P. W. (2002). Multiple feedback mechanisms activating

corticotropin-releasing hormone system in the brain during stress. Pharmacology

Biochemistry and Behavior, 73(1), 147–158. doi:10.1016/S0091-3057(02)00791-8

Mangiola, A., Vigo, V., Anile, C., Bonis, P. De, Marziali, G., & Lofrese, G. (2015). Role and

Importance of IGF-1 in Traumatic Brain Injuries, 2015.

87

Markham, J. a., & Koenig, J. I. (2011). Prenatal stress: Role in psychotic and depressive

diseases. Psychopharmacology, 214(1), 89–106. doi:10.1007/s00213-010-2035-0

Mccarthy, M. M., Auger, A. P., Bale, T. L., Vries, G. J. De, Gregory, a, Forger, N. G., …

Wilson, M. E. (2010). The Epigenetics of Sex Differences in the Brain. Journal of

Neuroscience, 29(41), 12815–12823. doi:10.1523/JNEUROSCI.3331-09.2009.

McGill, B. E., Bundle, S. F., Yaylaoglu, M. B., Carson, J. P., Thaller, C., & Zoghbi, H. Y.

(2006). Enhanced anxiety and stress-induced corticosterone release are associated with

increased Crh expression in a mouse model of Rett syndrome. Proceedings of the

National Academy of Sciences of the United States of America, 103(48), 18267–18272.

doi:10.1073/pnas.0608702103

McKee, S., Grissom, N. M., Bowman, N., & Reyes, T. M. (2014, November). Perinatal high fat

diet leads to DNMT1 deficits in the offspring prefrontal cortex: mRNA overexpression,

reduced activity and cytoplasmic sequestration. Poster presented at the Neuroscience

2014 Conference of Society for Neuroscience, Washington, DC.

McGowan, P. O., Sasaki, A., D’Alessio, A. C., Dymov, S, Labonté, B, & Szyf, M. E. A. (2010).

Epigenetic regulation of the glucocorticoid receptor in human brain associates with

childhood abuse. Nature Neuroscience, 12(3), 342–348. doi:10.1038/nn.2270.Epigenetic

Milanski, M., Degasperi, G., Coope, a., Morari, J., Denis, R., Cintra, D. E., … Velloso, L. a.

(2009). Saturated Fatty Acids Produce an Inflammatory Response Predominantly through

the Activation of TLR4 Signaling in Hypothalamus: Implications for the Pathogenesis of

Obesity. Journal of Neuroscience, 29(2), 359–370. doi:10.1523/JNEUROSCI.2760-

08.2009

88

Mikaelsson, M. A., Constância, M., Dent, C. L., Wilkinson, L. S., & Humby, T. (2013).

Placental programming of anxiety in adulthood revealed by Igf2-null models. Nature

Communications, 4(May), 2311. doi:10.1038/ncomms3311

Mitra, R., Ferguson, D., & Sapolsky, R. M. (2009). Mineralocorticoid Receptor Overexpression

in Basolateral Amygdala Reduces Corticosterone Secretion and Anxiety. Biological

Psychiatry, 66(7), 686–690. doi:10.1016/j.biopsych.2009.04.016

Mitra, R., & Sapolsky, R. M. (2008). Acute corticosterone treatment is sufficient to induce

anxiety and amygdaloid dendritic hypertrophy. Proceedings of the National Academy of

Sciences of the United States of America, 105(14), 5573–5578.

doi:10.1073/pnas.0705615105

Mueller, B. R., & Bale, T. L. (2008). Sex-specific programming of offspring emotionality after

stress early in pregnancy. The Journal of Neuroscience : The Official Journal of the

Society for Neuroscience, 28(36), 9055–9065. doi:10.1523/JNEUROSCI.1424-08.2008

Munhoz, C. D., Lepsch, L. B., Kawamoto, E. M., Malta, M. B., Lima Lde, S., Avellar, M. C.,

Sapolsky, R. M., & Scavone, C. (2006). Chronic unpredictable stress exacerbates

lipopolysaccharide-induced activation of nuclear factor- kappaB in the frontal cortex and

hippocampus via glucocorticoid secretion. Journal of Neuroscience, 26(14), 3813–3820.

doi: 10.1523/JNEUROSCI.4398-05.2006

Munhoz, C. D., Sorrells S. F., Caso J. R., Scavone C., & Sapolsky, R.M. (2010). Glucocorticoids

exacerbate lipopolysaccharide-induced signaling in the frontal cortex and hippocampus in

a dose-dependent manner. Journal of Neuroscience, 30(41), 13690–13698. doi:

10.1523/JNEUROSCI.0303-09.2010

89

Murphy, T. M., Donovan, A. O., Mullins, N., Farrelly, C. O., McCann, A., Malone, K., …

Malone, K. (2014). Anxiety is associated with higher levels of global DNA methylation

and altered expression of epigenetic and interleukin-6 genes. Psychiatric Genetics,

(August 2015), 1–8. doi:10.1097/YPG.0000000000000055

Naef, L., Gratton, A., & Walker, C.-D. (2013). Exposure to high fat during early development

impairs adaptations in dopamine and neuroendocrine responses to repeated stress. Stress

(Amsterdam, Netherlands), 3890(5), 1–9. doi:10.3109/10253890.2013.805321

Nair, A., & Bonneau, R.H. (2006). Stress-induced elevation of glucocorticoids increases

microglia proliferation through NMDA receptor activation. Journal of

Neuroimmunology, 171, 72–85. doi:10.1016/j.jneuroim.2005.09.012

Najjar, S., Pearlman, D. M., Alper, K., Najjar, A., & Devinsky, O. (2013). Neuroinflammation

and psychiatric illness. Journal of Neuroinflammation, 10, 1–24. doi:10.1186/1742-2094-

10-43

Nawa, H., & Takei, N. (2006). Recent progress in animal modeling of immune inflammatory

processes in schizophrenia: Implication of specific cytokines. Neuroscience Research.

doi:10.1016/j.neures.2006.06.002

Nelson, G., Wilde, G. J. C., Spiller, D. G., Kennedy, S. M., Ray, D. W., Sullivan, E., … White,

M. R. H. (2003). NF-kappaB signaling is inhibited by glucocorticoid receptor and STAT6

via distinct mechanisms. Journal of Cell Science, 116(Pt 12), 2495–503.

doi:10.1242/jcs.00461

90

Niehrs, C., & Schäfer, A. (2012). Active DNA demethylation by Gadd45 and DNA repair.

Trends in Cell Biology, 22(4), 220–227. doi:10.1016/j.tcb.2012.01.002

Nugent, B. M., Wright, C. L., Shetty, A. C., Hodes, G. E., Lenz, K. M., Mahurkar, A., …

Mccarthy, M. M. (2015). Brain feminization requires active repression of masculinization

via DNA methylation. Nature Neuroscience, 18(August 2014). doi:10.1038/nn.3988

O'Connor, T. G. (2002). Maternal antenatal anxiety and children’s behavioural/emotional

problems at 4 years: Report from the Avon Longitudinal Study of Parents and Children.

The British Journal of Psychiatry, 180(6), 502–508. doi:10.1192/bjp.180.6.502

O’Donovan, A., Hughes, B. M., Slavich, G. M., Lynch, L., Cronin, M.-T., O’Farrelly, C., &

Malone, K. M. (2010). Clinical anxiety, cortisol and interleukin-6: Evidence for

specificity in emotion–biology relationships. Brain, Behavior, and Immunity, 24(7),

1074–1077. doi:10.1016/j.bbi.2010.03.003

Oberlander, T. F., Weinberg, J., Papsdorf, M., Grunau, R., Misri, S., & Devlin, A. M. (2008).

Prenatal exposure to maternal depression, neonatal methylation of human glucocorticoid

receptor gene (NR3C1) and infant cortisol stress responses. Epigenetics, 3(2), 97–106.

doi:10.4161/epi.3.2.6034

Ong, Z. Y., Muhlhausler, B. S. (2011). Maternal ‘‘junk-food’’ feeding of rat dams alters food

choices and development of the mesolimbic reward pathway in the offspring. The FASEB

Journal : Official Publication of the Federation of American Societies for Experimental

Biology, 25(7), 2167–2179. doi: 10.1096/fj.10-178392

91

Peleg-Raibstein, D., Luca, E., & Wolfrum, C. (2012). Maternal high-fat diet in mice programs

emotional behavior in adulthood. Behavioural Brain Research, 233(2), 398–404.

doi:10.1016/j.bbr.2012.05.027

Pham, T. D., MacLennan, N. K., Chiu, C. T., Laksana, G. S., Hsu, J. L., & Lane, R. H. (2003).

Uteroplacental insufficiency increases apoptosis and alters p53 gene methylation in the

full-term IUGR rat kidney. American Journal of Physiology. Regulatory, Integrative and

Comparative Physiology, 285(5), R962–R970. doi:10.1152/ajpregu.00201.2003

Pilsner, J. R., Lazarus, A. L., Nam, D. H., Letcher, R. J., Sonne, C., Dietz, R., & Basu, N. (2010).

Mercury-associated DNA hypomethylation in polar bear brains via the LUminometric

Methylation Assay: A sensitive method to study epigenetics in wildlife. Molecular

Ecology, 19(2), 307–314. doi:10.1111/j.1365-294X.2009.04452.x

Postal, M., Costallat, L. T. L., & Appenzeller, S. (2011). Neuropsychiatric manifestations in

systemic lupus erythematosus: epidemiology, pathophysiology and management. CNS

Drugs, 25(9), 721–36. doi:10.2165/11591670-000000000-00000

Purcell, R. H., Sun, B., Pass, L. L., Power, M. L., Moran, T. H., & Tamashiro, K. L. (2011)

Maternal stress and high-fat diet effect on maternal behavior, milk composition, and pup

ingestive behavior. Physiology & Behavior, 104(3), 474–479. doi:

10.1016/j.physbeh.2011.05.012

Qin, C., Greenwood-Van Meerveld, B., & Foreman, R. D. (2003). Spinal neuronal responses to

urinary bladder stimulation in rats with corticosterone or aldosterone onto the amygdala.

Journal of Neurophysiology, 90(4), 2180–2189. doi:10.1152/jn.00298.2003

92

Quan, N., He, L., Lai, W., Shen, T., & Herkenham, M. (2000). Induction of IkBa mRNA

Expression in the Brain by Glucocorticoids : A Negative Feedback Mechanism for

Immune-to-Brain Signaling, 20(17), 6473–6477.

Ramamoorthy, S., & Cidlowski, J. a. (2013). Ligand-Induced Repression of the Glucocorticoid

Receptor Gene Is Mediated by an NCoR1 Repression Complex Formed by Long-Range

Chromatin Interactions with Intragenic Glucocorticoid Response Elements. Molecular

and Cellular Biology, 33(9), 1711–1722. doi:10.1128/MCB.01151-12

Ratnayake, U., Quinn, T., Walker, D. W., & Dickinson, H. (2013). Cytokines and the

neurodevelopmental basis of mental illness. Frontiers in Neuroscience, 7(7 OCT), 180.

doi:10.3389/fnins.2013.00180

Reul, J. M. H. M., Gesing, A., Droste, S., Stec, I. S. M., Weber, A., Bachmann, C., … Linthorst,

A. C. E. (2000). The brain mineralocorticoid receptor: Greedy for ligand, mysterious in

function. European Journal of Pharmacology, 405(1-3), 235–249. doi:10.1016/S0014-

2999(00)00677-4

Rhen, T., & Cidlowski, J. a. (2005). Antiinflammatory action of glucocorticoids--new

mechanisms for old drugs. The New England Journal of Medicine, 353(16), 1711–1723.

doi:10.1056/NEJMra050541

Rivera, H. M., Christiansen, K. J., & Sullivan, E. L. (2015). The role of maternal obesity in the

risk of neuropsychiatric disorders. Frontiers in Neuroscience, 9(June), 1–16.

doi:10.3389/fnins.2015.00194

93

Robson, P. J., Blannin, A. K., Walsh, N. P., Castell, L. M., & Gleeson, M. (1999). Effects of

exercise intensity, duration and recovery on in vitro neutrophil function in male athletes.

International Journal of Sports Medicine, 20(2), 128–35. doi:10.1055/s-2007-971106

Rodriguez, A. (2010). Maternal pre-pregnancy obesity and risk for inattention and negative

emotionality in children. Journal of Child Psychology and Psychiatry, and Allied

Disciplines, 51(2), 134–143. doi: 10.1111/j.1469-7610.2009.02133.x

Rohleder, N., Schommer, N. C., Hellhammer, D. H., Engel, R., & Kirschbaum, C. (2001). Sex

differences in glucocorticoid sensitivity of proinflammatory cytokine production after

psychosocial stress. Psychosomatic Medicine, 63(6), 966–72. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/11719636

Rozeboom, A. M., Akil, H., & Seasholtz, A. F. (2007). Mineralocorticoid receptor

overexpression in forebrain decreases anxiety-like behavior and alters the stress response

in mice. Proceedings of the National Academy of Sciences of the United States of

America, 104(11), 4688–4693. doi:10.1073/pnas.0606067104

Samaco, R. C., Mandel-Brehm, C., McGraw, C. M., Shaw, C. a, McGill, B. E., & Zoghbi, H. Y.

(2012). Crh and Oprm1 mediate anxiety-related behavior and social approach in a mouse

model of MECP2 duplication syndrome. Nature Genetics, 44(2), 206–211.

doi:10.1038/ng.1066

Saraiva, M., & O’Garra, A. (2010). The regulation of IL-10 production by immune cells. Nature

Reviews. Immunology, 10(3), 170–181. doi:10.1038/nri2711

94

Sarrazin, N., Di Blasi, F., Roullot-Lacarrière, V., Rougé-Pont, F., Le Roux, A., Costet, P., …

Piazza, P. V. (2009). Transcriptional effects of glucocorticoid receptors in the dentate

gyrus increase anxiety-related behaviors. PloS One, 4(11), e7704.

doi:10.1371/journal.pone.0007704

Sasaki, A., de Vega, W. C., St-Cyr, S., Pan, P., & McGowan, P. O. (2013). Perinatal high fat diet

alters glucocorticoid signaling and anxiety behavior in adulthood. Neuroscience, 240, 1–

12. doi:10.1016/j.neuroscience.2013.02.044

Sasaki, A., Erb, S., & McGowan, P. O. (2015). The effect of maternal overnutrition on reward

and anxiety in offspring. Manuscript submitted for publication.

Schaefer, C. a, Brown, a S., Wyatt, R. J., Kline, J., Begg, M. D., Bresnahan, M. a, & Susser, E.

S. (2000). Maternal prepregnant body mass and risk of schizophrenia in adult offspring.

Schizophrenia Bulletin, 26(2), 275–86. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/10885630

Schmitt, A., Malchow, B., Hasan, A., & Falkai, P. (2014). The impact of environmental factors

in severe psychiatric disorders. Frontiers in Neuroscience, 8(February), 1–10.

doi:10.3389/fnins.2014.00019

Schwarz, J. M., Hutchinson, M. R., & Bilbo, S. D. (2011). Early-Life Experience Decreases

Drug-Induced Reinstatement of Morphine CPP in Adulthood via Microglial-Specific

Epigenetic Programming of Anti-Inflammatory IL-10 Expression. Journal of

Neuroscience, 31(49), 17835–17847. doi:10.1523/JNEUROSCI.3297-11.2011

95

Scotland, R. S., Stables, M. J., Madalli, S., Watson, P., & Gilroy, D. W. (2011). Sex differences

in resident immune cell phenotype underlie more efficient acute inflammatory responses

in female mice. Blood, 118(22), 5918–27. doi:10.1182/blood-2011-03-340281

Shapero, B. G., Black, S. K., Liu, R. T., Klugman, J., Bender, R. E., Abramson, L. Y., & Alloy,

L. B. (2014). Stressful life events and depression symptoms: the effect of childhood

emotional abuse on stress reactivity. Journal of Clinical Psychology, 70(3), 209–23.

doi:10.1002/jclp.22011

Sharma, D., Bhave, S., Gregg, E., & Uht, R. (2013). Dexamethasone induces a putative repressor

complex and chromatin modifications in the CRH promoter. Molecular Endocrinology

(Baltimore, Md.), 27(November), 1142–52. doi:10.1210/me.2013-1079

Simmons, R. (2008). Perinatal programming of obesity. Seminars in Perinatology, 32(5), 371–4.

doi:10.1053/j.semperi.2008.08.004

Smith, E. M., Cadet, P., Stefano, G. B., Opp, M. R., & Hughes, T. K. Jr. (1999). IL-10 as a

mediator in the HPA axis and brain. Journal of Neuroimmunology, 100, 140-8.

Smith, S. M., & Vale, W. W. (2006). The role of the hypothalamic-pituitary-adrenal axis in

neuroendocrine responses to stress. Dialogues in Clinical Neuroscience, 8(4), 383–395.

doi:10.1038/nrendo.2011.222

Smythe, J. W., Murphy, D., Timothy, C., & Costall, B. (1997). Hippocampal mineralocorticoid,

but not glucocorticoid, receptors modulate anxiety-like behavior in rats. Pharmacology,

Biochemistry, and Behavior, 56(3), 507–513.

96

Sorrells, S. F., Caso, J. R., Munhoz, C. D., & Sapolsky, R. M. (2009). The stressed CNS: when

glucocorticoids aggravate inflammation. Neuron, 64(1), 33–9.

doi:10.1016/j.neuron.2009.09.032

Stein-Behrens, B., Mattson, M. P., Chang, I., Yeh, M., & Sapolsky, R.. (1994) Stress exacerbates

neuron loss and cytoskeletal pathology in the hippocampus. Journal of Neuroscience, 14,

5373–5380.

Strohle, A., & Holsboer, F. (2003). Stress responsive neurohormones in depression and anxiety.

Pharmacopsychiatry, 36 Suppl 3, S207–14. doi:10.1055/s-2003-45132

Sullivan, E. L., Grayson, B., Takahashi, D., Robertson, N., Maier, A., Bethea, C. L., Smith,

M.S., Coleman, K., & Grove, K. L. (2010) Chronic consumption of a high-fat diet during

pregnancy causes perturbations in the serotonergic system and increased anxiety-like

behavior in nonhuman primate offspring. Journal of Neuroscience, 30(10), 3826–30. doi:

10.1523/JNEUROSCI.5560-09.2010

Sullivan, E. L., Riper, K. M., Lockard, R., & Valleau, J. C. (2015). Maternal high-fat diet

programming of the neuroendocrine system and behavior. Hormones and Behavior.

doi:10.1016/j.yhbeh.2015.04.008

Suzuki, M. M., & Bird, A. (2008). DNA methylation landscapes: provocative insights from

epigenomics. Nature Reviews. Genetics, 9(6), 465–76. doi:10.1038/nrg2341

Svennerholm, L., & Vanier, M. T. (1978). Lipid and fatty acid composition of human cerebral

myelin during development. Advances in experimental medicine and biology, 100, 27–41.

Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/696475

97

Tamashiro, K. L., Terrillion, C. E., Hyun, J., Koenig, J. I., & Moran, T. H. (2009). Prenatal stress

or high-fat diet increases susceptibility to diet- induced obesity in rat offspring. Diabetes,

58(5), 1116–1125. doi: 10.2337/db08-1129

Teicher, M. H., & Samson, J. A. (2013). Childhood Maltreatment and Psychopathology: A Case

for Ecophenotypic Variants as Clinically and Neurobiologically Distinct Subtypes.

American Journal of Psychiatry, 170(10), 1114. doi:10.1176/appi.ajp.2013.12070957

Ter Heegde, F., De Rijk, R. H., & Vinkers, C. H. (2014). The brain mineralocorticoid receptor

and stress resilience. Psychoneuroendocrinology, 52, 92–110.

doi:10.1016/j.psyneuen.2014.10.022

Theisen, J. W. M., Gucwa, J. S., Yusufzai, T., Khuong, M. T., & Kadonaga, J. T. (2013).

Biochemical analysis of histone deacetylase-independent transcriptional repression by

MeCP2. The Journal of Biological Chemistry, 288(10), 7096–104.

doi:10.1074/jbc.M112.438697

Tronche, F., Kellendonk, C., Kretz, O., Gass, P., Anlag, K., Orban, P. C., … Schütz, G. (1999).

Disruption of the glucocorticoid receptor gene in the nervous system results in reduced

anxiety. Nature Genetics, 23(1), 99–103. doi:10.1038/12703

Tu, H., Rady, P. L., Juelich, T., Tyring, S. K., Koldzic-Zivanovic, N., Smith, E. M., & Hughes,

T. K. (2007). Interleukin-10 regulated gene expression in cells of hypothalamic-pituitary-

adrenal axis origin. Cellular and Molecular Neurobiology, 27(2), 161–170.

doi:10.1007/s10571-006-9137-6

98

Urb, M., Anier, K., Matsalu, T., Aonurm-Helm, A., & Timmusk, T. (2015). Corticosterone

induces DNA methyltransferases expression in rat cortical neurons. SpringerPlus,

4(Suppl 1), P50. doi:10.1186/2193-1801-4-S1-P50

Van den Bergh, B. R. H., Mennes, M., Stevens, V., Van Der Meere, J., Börger, N., Stiers, P., …

Lagae, L. (2006). ADHD deficit as measured in adolescent boys with a continuous

performance task is related to antenatal maternal anxiety. Pediatric Research, 59(1), 78–

82. doi:10.1203/01.pdr.0000191143.75673.52

Van Lieshout, R. J., Robinson, M., & Boyle, M. H. (2013). Maternal pre-pregnancy body mass

index and internalizing and externalizing problems in offspring. Canadian Journal of

Psychiatry. Revue Canadienne de Psychiatrie, 58(3), 151–9. Retrieved from

http://www.ncbi.nlm.nih.gov/pubmed/23461886

Van Winkel, R., Stefanis, N. C., & Myin-Germeys, I. (2008). Psychosocial stress and psychosis.

A review of the neurobiological mechanisms and the evidence for gene-stress interaction.

Schizophrenia Bulletin, 34(6), 1095–105. doi:10.1093/schbul/sbn101

Vandevyver, S., Dejager, L., Van Bogaert, T., Kleyman, A., Liu, Y., Tuckermann, J., & Libert,

C. (2012). Glucocorticoid receptor dimerization induces MKP1 to protect against TNF-

induced inflammation. Journal of Clinical Investigation, 122(6), 2130–2140.

doi:10.1172/JCI60006

Varese, F., Smeets, F., Drukker, M., Lieverse, R., Lataster, T., Viechtbauer, W., … Bentall, R. P.

(2012). Childhood Adversities Increase the Risk of Psychosis: A Meta-analysis of

Patient-Control, Prospective- and Cross-sectional Cohort Studies. Schizophrenia Bulletin,

38(4), 661–671. doi:10.1093/schbul/sbs050

99

Voorhees, J. L., Tarr, A. J., Wohleb, E. S., Godbout, J. P., Mo, X., Sheridan, J. F., … Marsh, C.

B. (2013). Prolonged Restraint Stress Increases IL-6, Reduces IL-10, and Causes

Persistent Depressive-Like Behavior That Is Reversed by Recombinant IL-10. PLoS

ONE, 8(3). doi:10.1371/journal.pone.0058488

Wang, X., Athayde, N., & Trudinger, B. (2003). A proinflammatory cytokine response is present

in the fetal placental vasculature in placental insufficiency. American Journal of

Obstetrics and Gynecology, 189(5), 1445–1451. doi:10.1067/S0002-9378(03)00652-5

Weaver, I. C. G., Cervoni, N., Champagne, F. a, D’Alessio, A. C., Sharma, S., Seckl, J. R., …

Meaney, M. J. (2004). Epigenetic programming by maternal behavior. Nature

Neuroscience, 7(8), 847–854. doi:10.1038/nn1276

Weaver, I. C. G., D’Alessio, A. C., Brown, S. E., Hellstrom, I. C., Dymov, S., Sharma, S., …

Meaney, M. J. (2007). The transcription factor nerve growth factor-inducible protein a

mediates epigenetic programming: altering epigenetic marks by immediate-early genes.

The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 27(7),

1756–1768. doi:10.1523/JNEUROSCI.4164-06.2007

Weinstock, M. (2005). The potential influence of maternal stress hormones on development and

mental health of the offspring. Brain, Behavior, and Immunity, 19(4), 296–308.

doi:10.1016/j.bbi.2004.09.006

Weinstock, M. (2008). The long-term behavioural consequences of prenatal stress. Neuroscience

and Biobehavioral Reviews, 32(6), 1073–86. doi:10.1016/j.neubiorev.2008.03.002

100

Welberg, L. a M., Seckl, J. R., & Holmes, M. C. (2000). Inhibition of 11β-hydroxysteroid

dehydrogenase, the foetoplacental barrier to maternal glucocorticoids, permanently

programs amygdala GR mRNA expression and anxiety-like behaviour in the offspring.

European Journal of Neuroscience, 12(3), 1047–1054. doi:10.1046/j.1460-

9568.2000.00958.x

Welberg, L. a M., Seckl, J. R., & Holmes, M. C. (2001). Prenatal glucocorticoid programming of

brain corticosteroid receptors and corticotrophin-releasing hormone: possible

implications for behaviour. Neuroscience, 104(1), 71–79. doi:10.1016/S0306-

4522(01)00065-3

White, C. L., Pistell, P. J., Purpera, M. N., Gupta, S., Fernandez-Kim, S.-O., Hise, T. L., …

Bruce-Keller, A. J. (2009). Effects of high fat diet on Morris maze performance,

oxidative stress, and inflammation in rats: contributions of maternal diet. Neurobiology of

Disease, 35(1), 3–13. doi:10.1016/j.nbd.2009.04.002

Williamson, L. L., Sholar, P. W., Mistry, R. S., Smith, S. H., & Bilbo, S. D. (2011). Microglia

and Memory: Modulation by Early-life Infection. The Journal of Neuroscience : The

Official Journal of the Society for Neuroscience, 31(43), 15511–15521.

doi:10.1523/JNEUROSCI.3688-11.2011

Zaretsky, M. V, Alexander, J. M., Byrd, W., & Bawdon, R. E. (2004). Transfer of inflammatory

cytokines across the placenta. Obstetrics and Gynecology, 103(3), 546–550.

doi:10.1097/01.AOG.0000114980.40445.83

Ziaja, M. (2013). Septic Encephalopathy. Current Neurology and Neuroscience Reports, 13(10),

383. doi:10.1007/s11910-013-0383-y

101

Appendix

Supplementary Table 1. Transcriptional response to CORT challenge in the amygdala (post-

hocs). Fisher's PLSD post-hocs of qPCR gene expression data separated by (a) males and (b)

females. CORT-induced anti-inflammatory signaling is exacerbated in Perinatal HFD-exposed

groups. # p < 0.1, * p < 0.05, ** p < 0.01. Shaded boxes: Diet x Drug interaction p < 0.05.

a) Males

b) Females

Function Gene Effect of CORT within diet group Effect of diet within drug group

GC signalling GR CHD CTL > CHD CORT ** HFD CORT > CHD CORT *

MR CHD CTL > CHD CORT * HFD CTL < CHD CTL #

CRH HFD CTL < HFD CORT # HFD CORT > CHD CORT *

Pro-inflammatory CD11b HFD CTL < CHD CTL #

NFkB CTL > CORT, HFD + CHD #

IL-6 CTL < CORT, HFD + CHD **

IL-6/IL-10 CTL < CORT, HFD + CHD **

Anti-inflammatory IGF-1 CHD CTL > CHD CORT # HFD CORT > CHD CORT #

IkBa CTL < CORT, HFD + CHD ** HFD CORT > CHD CORT **

MKP-1 HFD CTL < HFD CORT ** HFD CORT > CHD CORT *

IL-10 CHD CTL > CHD CORT #

Epigenetic regulation DNMT1 CHD CTL > CHD CORT * HFD CORT > CHD CORT **

DNMT3a CTL > CORT, HFD + CHD **

DNMT3b

MeCP2 CHD CTL > CHD CORT # HFD CORT > CHD CORT *

Gadd45b HFD CTL < HFD CORT # HFD CORT > CHD CORT #

Function Gene Effect of CORT within diet group Effect of diet within drug group

GC signalling GR HFD CTL > HFD CORT *

MR

CRH

Pro-inflammatory CD11b

NFkB

IL-6

IL-6/IL-10 HFD CTL < HFD CORT *

Anti-inflammatory IGF-1

IkBa HFD CTL < HFD CORT *

MKP-1

IL-10

Epigenetic regulation DNMT1

DNMT3a

DNMT3b

MeCP2

Gadd45b

102

Supplementary Table 2. Transcriptional response to CORT challenge in the hippocampus

(post-hocs). Fisher's PLSD post-hocs of qPCR gene expression data separated by (a) males and

(b) females. CORT-induced anti-inflammatory signaling is exacerbated in Perinatal HFD-

exposed groups. # p < 0.1, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Shaded

boxes: Diet x Drug interaction p < 0.05.

a) Males

b) Females

Function Gene Effect of CORT within diet group Effect of diet within drug group

GC signalling GR CHD CTL > CHD CORT * HFD CORT > CHD CORT *

MR CHD CTL > CHD CORT * HFD CORT > CHD CORT #

Pro-inflammatory CD11b

NFkB

IL-6 CTL < CORT, CHD ***, HFD * HFD CTL > CHD CTL #

IL-6/IL-10 CHD CTL < CHD CORT **

Anti-inflammatory IGF-1

IkBa CTL < CORT, CHD ****, HFD ** HFD CORT > CHD CORT *

MKP-1 CTL < CORT, CHD ****, HFD ** HFD CORT > CHD CORT *

IL-10

Epigenetic regulation DNMT1

DNMT3a

MeCP2

Gadd45b

Function Gene Effect of CORT within diet group Effect of diet within drug group

GC signalling GR CHD CTL > CHD CORT ** HFD CTL < CHD CTL *

MR

Pro-inflammatory CD11b CHD CTL > CHD CORT ** HFD CTL < CHD CTL *

NFkB CHD CTL > CHD CORT ** HFD CTL < CHD CTL *

IL-6 HFD CTL < HFD CORT #

IL-6/IL-10 HFD CTL < HFD CORT #

Anti-inflammatory IGF-1

IkBa HFD CTL < HFD CORT ** HFD CTL < CHD CTL, HFD CORT > CHD CORT *

MKP-1 CHD CTL > CHD CORT *, HFD CTL < HFD CORT # HFD CTL < CHD CTL, HFD CORT > CHD CORT #

IL-10

Epigenetic regulation DNMT1 CHD CTL > CHD CORT **

DNMT3a CHD CTL > CHD CORT, CHD **, HFD #

MeCP2 CHD CTL > CHD CORT, CHD ***, HFD * HFD CORT > CHD CORT #

Gadd45b CHD CTL > CHD CORT *

103

Supplementary Table 3. Transcriptional response to LPS challenge in the amygdala (post-

hocs). Fisher's PLSD post-hocs of qPCR gene expression data separated by (a) males and (b)

females. LPS-induced anti-inflammatory signaling is exacerbated in perinatal HFD-exposed

groups. # p < 0.1, * p < 0.05, ** p < 0.01, *** p < 0.001.

a) Males

b) Females

Function Gene Effect of LPS within diet group Effect of diet within drug group

GC signalling GR

MR CHD CTL > CHD LPS #

Pro-inflammatory CD11b HFD CTL < CHD CTL *

NFkB

IL-6 CTL < LPS, HFD + CHD *

IL-6/IL-10 CTL < LPS, CHD **, HFD *

Anti-inflammatory IGF-1 CHD CTL < CHD LPS *

IkBa CTL < LPS, HFD **, CHD *

MKP-1 HFD CTL < HFD LPS *

IL-10 CTL > LPS, HFD ***, CHD *

Function Gene Effect of LPS within diet group Effect of diet within drug group

GC signalling GR CHD CTL > CHD LPS #

MR CTL < LPS, HFD + CHD #

Pro-inflammatory CD11b CHD CTL > CHD LPS *

NFkB HFD LPS < CHD LPS *

IL-6 CTL < LPS, HFD **, CHD #

IL-6/IL-10 CTL < LPS, HFD + CHD **

Anti-inflammatory IGF-1

IkBa HFD CTL < HFD LPS *

MKP-1

IL-10

104

Supplementary Table 4. Transcriptional response to CORT/LPS challenge in the amygdala

(post-hocs). Fisher's PLSD post-hocs of qPCR gene expression data separated by (a) males and

(b) females. CORT/LPS-induced pro-inflammatory response is altered in Perinatal HFD-exposed

groups. # p < 0.1, * p < 0.05, ** p < 0.01, *** p < 0.001. Shaded boxes: Diet x Drug interaction

p < 0.05.

a) Males

b) Females

Function Gene Effect of CORT/LPS within diet group Effect of diet within drug group

GC signalling GR CTL > CORT/LPS, HFD *, CHD ***

MR CHD CTL > CHD CORT/LPS ** HFD CTL < CHD CTL *

CRH CHD CTL < CHD CORT/LPS **

Pro-inflammatory CD11b

NFkB CTL > CORT/LPS, CHD ***, HFD * HFD CORT/LPS > CHD CORT/LPS *

IL-6 CTL < CORT/LPS, HFD ***, CHD **

IL-6/IL-10 CTL < CORT/LPS ** HFD CORT/LPS < CHD CORT/LPS #

Anti-inflammatory IGF-1 CTL > CORT/LPS, CHD **, HFD *

IkBa CTL < CORT/LPS, HFD + CHD **

MKP-1

IL-10 CTL > CORT/LPS, CHD **, HFD *

Epigenetic regulation DNMT1 CHD CTL > CHD CORT/LPS *

DNMT3a CHD CTL > CHD CORT/LPS #

DNMT3b

MeCP2

Gadd45b CHD CTL < CHD CORT/LPS **

Function Gene Effect of CORT/LPS within diet group Effect of diet within drug group

GC signalling GR CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS **

MR CTL > CORT/LPS, CHD **, HFD * HFD CORT/LPS < CHD CORT/LPS **

CRH CHD CTL < CHD CORT/LPS ** HFD CORT/LPS < CHD CORT/LPS **

Pro-inflammatory CD11b CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS *

NFkB CTL > CORT/LPS, HFD **, CHD * HFD CORT/LPS < CHD CORT/LPS #

IL-6

IL-6/IL-10 CTL < CORT/LPS, HFD + CHD ** HFD CORT/LPS < CHD CORT/LPS #

Anti-inflammatory IGF-1 CHD CTL > CHD CORT/LPS #

IkBa

MKP-1 HFD CTL > HFD CORT/LPS *

IL-10 CTL > CORT/LPS, HFD + CHD **

Epigenetic regulation DNMT1 HFD CTL > HFD CORT/LPS * HFD CORT/LPS < CHD CORT/LPS *

DNMT3a HFD CTL > HFD CORT/LPS # HFD CORT/LPS < CHD CORT/LPS *

DNMT3b HFD CTL > HFD CORT/LPS *

MeCP2

Gadd45b HFD CTL < CHD CTL #