Effects of developmental vitamin D deficiency and prenatal exposure to ethanol on brain development and behaviour in mice

Michelle Carolina Sanchez Vega Bachelor of Biomedical Science, First Class Honours (Neuroscience)

A thesis submitted for the degree of Doctor of Philosophy at The University of Queensland in September 2017 Queensland Brain Institute Abstract

There is a high prevalence of vitamin D deficiency and exposure to low levels of alcohol in pregnant women (Bodnar et al 2007, Daly et al 2012). However, there are a paucity of studies that have addressed the impact of both of these exposures on the offspring’s vulnerability to neuropsychological disorders later in life. The aim of this thesis was to examine whether the absence of vitamin D during gestation in mice would alter the effects of prenatal exposure to low dose ethanol on brain function and behaviour.

Four-week old female C57Bl/6J and BALB/c mice were placed on a developmental vitamin D deficient (DVD) or standard diet for 6 weeks and mated at 10 weeks of age. Females were exposed to either 10 %(v/v) ethanol or water between gestational days (GD) 0-8 and all were offered water thereafter. Behavioural analyses of the offspring included ultrasonic vocalizations (USV) at postnatal day (P) 7, social interaction (SI) at P21, locomotion and spatial learning and memory at P70. Ex-vivo MRI, next-generation sequencing (NGS) and proteomics assays were carried out in neonatal brains, followed by expression analyses on potential candidates in P0, P21 and P70 tissue.

MRI revealed no gross brain anatomy in DVD-deficient or prenatal ethanol exposure (PEE) P0 samples, however, detailed histological analysis revealed a main effect of Diet on hippocampal volume at birth. RNA sequencing showed that pathways such as cell growth, cell morphology as well as cell death and survival were affected as a result of both DVD deficiency and PEE, whereby a greater number of transcripts were affected in the combination group versus individual factors. This was not observed in proteomics data, suggesting the combined effects of DVD-PEE was more prominent at mRNA level and did not translate to altered levels of . analysis on transcripts involved in dopamine regulation and neurodevelopmental markers via qRT-PCR, FoxP2 and Darpp-32 were downregulated as a main effect of PEE at P0, while there was a main effect of PEE on Darpp-32 and a main effect of Diet on D2R at P21. The main findings at P7-21 were increased USV calling rate in PEE males, altered distribution of call types among DVD and PEE groups and impaired SI in PEE males. Hypolocomotion was encountered in PEE adult males, while learning and memory performance in the APA task was unaffected in all Groups. Vglut2 was downregulated in PEE adult male . The combination of DVD-PEE did not exacerbate the alterations resulting from DVD or PEE, rather it appears as though these exposures acted on independent pathways. Taken

ii together, the current results indicate that DVD deficiency temporally increased the offspring vulnerability to PEE with a clear effect on disrupting neonatal gene expression. However, these effects were transient and dissipated during postnatal development suggesting that there was a critical window of vulnerability to these exposures. Despite the limited evidence to support the interaction of DVD and PEE during the postnatal period, males were more vulnerable than female offspring to the detrimental effects of PEE. Therefore, based on these studies in mice we suggest that maintenance of optimal vitamin D levels and abstinence from alcohol during pregnancy would reduce risk of later disruption to brain function and behaviour in the offspring.

iii Declaration by Author

This thesis is composed of my original work, and contains no material previously published or written by another person except where due reference has been made in the text. I have clearly stated the contribution by others to jointly-authored works that I have included in my thesis.

I have clearly stated the contribution of others to my thesis as a whole, including statistical assistance, survey design, data analysis, significant technical procedures, professional editorial advice, and any other original research work used or reported in my thesis. The content of my thesis is the result of work I have carried out since the commencement of my research higher degree candidature and does not include a substantial part of work that has been submitted to qualify for the award of any other degree or diploma in any university or other tertiary institution. I have clearly stated which parts of my thesis, if any, have been submitted to qualify for another award.

I acknowledge that an electronic copy of my thesis must be lodged with the University Library and, subject to the policy and procedures of The University of Queensland, the thesis be made available for research and study in accordance with the Copyright Act 1968 unless a period of embargo has been approved by the Dean of the Graduate School.

I acknowledge that copyright of all material contained in my thesis resides with the copyright holder(s) of that material. Where appropriate I have obtained copyright permission from the copyright holder to reproduce material in this thesis.

iv Publications during candidature

Peer-reviewed publications

Zhang, C.R., Ho, M.F., Sanchez Vega, M.C., Burne, T.H.J., Chong, S. (2015) Prenatal ethanol exposure alters adult hippocampal VGLUT2 expression with concomitant changes in DNA methylation, H3K4 trimethylation and miR-467b-5p levels. Epigenetics & chromatin 8, 40.

Conference Abstracts

Sanchez Vega, M., Chong, S., and Burne, T.H.J. (2016). Developmental vitamin D deficiency ameliorates the effects of prenatal exposure to ethanol. Proceedings of the Society for Neuroscience. 34, 101. Poster Presentation.

Sanchez Vega, M., Chong, S., and Burne, T.H.J. (2015). Does developmental vitamin D deficiency exacerbate the effects of second hit exposures?. Proceedings of the International Society for Neurochemistry. 34, 101. Poster Presentation.

Sanchez Vega, M., Chong, S., and Burne, T.H.J. (2014). Early gestational exposure to moderate concentrations of ethanol leads to an age-dependent decline in attentional processing in C57Bl/6J mice. Proceedings of the Australian Neuroscience Society. 34, 101. Poster Presentation.

Publications included in this thesis

No publications

v

Contribution by others to the thesis

Associate Professor Thomas Burne was my principle supervisor and provided support in designing and analysing each experiment. Dr Suyinn Chong also provided feedback on thesis drafts. Dr Nyoman Kurniawan from the Centre of Advanced Imaging, The University of Queensland, aided in the magnetic resonance imaging in Chapter 4. Dr Robert Sullivan from the Queensland Brain Institute, The University of Queensland, aided in the preparation of histology samples in Chapter 4. Kara Jaeschke and Henry Simila provided research support to conduct the alcohol metabolism assays in Chapter 3. Regina Yu conducted part of qRT-PCR in Chapter 5 as part of an undergraduate research project. Kyna-Anne Conn and Dana Bradford conducted immunohistochemistry staining and Emily Hartl conducted protein localization analysis included in Chapter 5 as part of an undergraduate research project.

Statement of parts of the thesis submitted to qualify for the award of another degree

None

vi Acknowledgements

I would like to express my gratitude to everyone that has helped along with this challenge. First and foremost, to my principal supervisor, Associate Professor Tom Burne, for the constant support, motivation, patience and enthusiasm. Thank you for pushing me to be better every day, with readily available metaphors and phrases of encouragement. You have taught me that science is not about finding the truth, rather about the journey of an idea and how to test it. You have shown me that although daunting at times, science is also exciting and worth waking up for every day. I would also like to thank my associate supervisor, Dr Suyinn Chong, for always being available to discuss new ideas, experiments and providing insightful comments throughout my candidature.

To all members in the lab, past and present, thank you for welcoming me and making this a great experience. Special thanks to Pauline Ko and Henry Simila, who seamlessly manage our lab and were always there to help; also to Suzy Alexander for her diligent planification and help with animal orders.

There are many people to thank at QBI that helped make these experiments happen, including staff from the animal house, histology and IT. To Kim Woolley, for her patience and willingness to help with animal procedures; Dr Rob Sullivan for his guidance on histology experiments; Dr Nyoman Kurniawan for his contribution to the processing and imaging of MRI samples; Dr Dana Bradford, Kyna Conn and Emily Hartl for their help with immunohisotchemistry; and my friend Dr Lachlan Harris, for his help in re-analysing and understanding my RNA-Seq data from a biological point of view. I would also like to acknowledge the financial support I obtained from the University of Queensland International scholarship, as well as the top-up scholarships I received from both QBI and the lab.

Finally, I would like to thank my family and friends who have supported me throughout my candidature. Dad, Mom and Grace, thank you for your relentless belief in me, even when I did not see myself capable; thank you for not letting me give up and being there in the good and bad times. To my friends back home, Mauri, Mene, Kendru, Baque and Mario, for making the distance feel irrelevant and always being just a message away. To my Aussie family, Ollie, Drew, Rochelle, Colty and Gottsy, for taking me in and keeping my life fun and exciting. To my fit friends, especially Tristan and Liam, for showing me the best

vii way to escape from stress, for keeping me healthy and strong, and more importantly, sane.

Last but not least, to Charlie, whom I could not have finished this without. Thank you for your everlasting amount of patience and words of encouragement, for being by my side when I could not even stand myself. For always keeping your cool when I lose mine and for making me smile every single day. I cannot thank you enough.

viii Keywords

Developmental vitamin D (DVD) deficiency, Prenatal Ethanol Exposure, brain development, animal model, C57Bl/6J mouse, BALB/c mouse, mouse behaviour, brain gene expression

Australian and New Zealand Standard Research Classifications (ANZSRC)

ANZSRC code: 110999, Neuroscience not elsewhere classified, 70% ANZSRC code: 060801, Animal Behaviour, 30%

Fields of Research (FoR) Classification

FoR code: 1109, Neurosciences, 100%

ix Dedication

To David.

x Table of Contents

List of Figures ...... xvi

List of Tables ...... xviii

List of Abbreviations ...... xix

Chapter 1. Literature review ...... 2 1.1 Environmental risk factors for neurodevelopmental disorders ...... 2 1.2 Developmental vitamin D deficiency as a risk factor for neurodevelopmental disorders ...... 4 1.3 Secondary insults to DVD deficiency ...... 9 1.4 Aims & outline of thesis ...... 13

Chapter 2. Establishing a two-hit model of DVD deficiency and PEE in C57BL/6J and BALB/c mice ...... 16 2.1 Introduction ...... 16 2.2 Material and methods ...... 19 2.2.1 Animals and housing ...... 19 2.2.2 Vitamin D deficiency ...... 20 2.2.3 Maternal ethanol exposure ...... 20 2.2.4 Behaviour ...... 21 2.2.4.1 Early developmental measures ...... 21 2.2.4.2 Adult locomotion ...... 21 2.2.5 Molecular ...... 21 2.2.5.1 Tissue collection ...... 21 2.2.5.2 Primer validation ...... 22 2.2.5.3 qRT-PCR ...... 22 2.2.6 Statistical analysis ...... 23 2.3 Results ...... 23 2.3.1 Breeding outcomes ...... 23 2.3.2 Developmental milestone assessment at P21 ...... 24 2.3.3 Adult locomotion (P70) ...... 27 2.3.4 Primer validation ...... 28 2.3.5 Vglut1-3 expression in early adulthood (P80-90) ...... 29 2.4 Discussion ...... 31 2.4.1 Effects of DVD deficiency and PEE on early development ...... 32 2.4.2 Effects of DVD deficiency and PEE on adult behaviour ...... 33 2.4.3 Prenatal exposures and gene expression ...... 33 2.4.4 Conclusion ...... 34

Chapter 3. Behavioural phenotype of adult C57BL/6J mice exposed to DVD deficiency and PEE...... 37 3.1 Introduction ...... 37 3.2 Materials and methods ...... 39 3.2.1 Animals, housing and exposures ...... 39 3.2.2 Adult locomotion ...... 39 3.2.3 Active place avoidance (APA) ...... 40 3.2.4 MK-801 locomotor response ...... 41 3.2.5 Statistical Analysis ...... 41 3.3 Results ...... 42 3.3.1 Adult locomotion ...... 42 3.3.2 Active place avoidance ...... 43 3.3.3 Psychomimetic-induced locomotion ...... 50 3.4 Discussion ...... 51 3.4.1 Effects of DVD deficiency and PEE on adult locomotion ...... 51 3.4.2 Effects of DVD deficiency and PEE on learning and memory ...... 52 3.4.3 Effects of DVD deficiency and PEE on psychomimetic-induced locomotion ...... 53 3.4.4 Conclusion ...... 54

Chapter 4. Interaction of DVD deficiency and PEE on maternal metabolism and neonatal gene and protein expression...... 56 4.1 Foreword ...... 56 4.2 Influence of maternal vitamin D status on alcohol metabolism ...... 56 4.2.1 Introduction ...... 56 4.2.2 Materials and methods ...... 58 4.2.2.1 Animals, housing and exposures ...... 58 4.2.2.2 Maternal blood sampling and tissue collection ...... 58 4.2.2.3 Maternal serum 25(OH)D levels ...... 58 4.2.2.4 Maternal blood alcohol concentration (BAC) determination ...... 58 4.2.2.5 Alcohol dehydrogenase (ADH) levels in maternal liver tissue...... 59 4.2.3 Results ...... 59 4.2.3.1 Maternal body weight and serum 25(OH)D levels ...... 59

xii 4.2.3.2 Maternal blood alcohol concentration ...... 60 4.2.3.3 Maternal liver Alcohol Dehydrogenase levels ...... 61 4.2.4 Discussion ...... 62 4.3 DVD deficiency and PEE on gene and protein expression ...... 63 4.3.1 Introduction ...... 63 4.3.2 Materials and methods ...... 64 4.3.2.1 Animals, housing and exposures ...... 64 4.3.2.2 RNA-sequencing ...... 65 a) Neonatal brain tissue collection ...... 65 b) Sample preparation ...... 65 c) Processing and analysis of RNA-Seq data ...... 65 4.3.2.3 SWATH Proteomics ...... 66 a) Sample preparation ...... 66 b) Strong cation exchange fractionation ...... 66 c) HPLC Triple-ToF 5600 mass spectrometer ...... 67 d) Protein Identification and quantification...... 67 e) Data processing and statistical analysis...... 68 4.3.2.4 Pathway analysis ...... 68 4.3.3 Results ...... 68 4.3.3.1 RNA-Sequencing ...... 68 4.3.3.2 SWATH Proteomics ...... 76 4.3.4 Discussion ...... 80 4.3.4.1 Effects of DVD deficiency and PEE on neonatal gene expression ...... 80 4.3.4.2 Effects of DVD deficiency and PEE on neonatal protein expression ...... 88 4.4 Conclusion ...... 92

Chapter 5. Effects of DVD deficiency and PEE on neonatal candidate gene expression and neuroanatomy...... 95 5.1 Introduction ...... 95 5.2 Materials and methods ...... 97 5.2.1 Animals, housing and exposures ...... 97 5.2.2 Neonatal brain tissue collection ...... 97 5.2.3 qRT-PCR ...... 97 5.2.4 Immunohistochemistry...... 98 a) Neonatal brain perfusions ...... 98 b) Tissue preparation for paraffin wax embedding ...... 98 xiii c) Wax-embedded tissue sectioning ...... 99 d) De-waxing and rehydration of wax-embedded tissue sections ...... 99 e) Antibodies and imaging ...... 99 5.2.5 Ex-vivo Magnetic Resonance Imaging (MRI) ...... 100 5.2.6 Histology ...... 100 5.2.7 Statistical analysis ...... 101 5.3 Results ...... 101 5.3.1 Gene expression of selected candidates (qRT-PCR) ...... 101 5.3.2 Immunohistochemistry...... 105 5.3.3 Ex-vivo MRI...... 106 5.3.4 Histology ...... 107 5.4 Discussion ...... 107 5.4.1 Effects of DVD deficiency and PEE on neonatal candidate gene expression .. 108 5.4.2 Effects of DVD deficiency and PEE on neonatal brain anatomy ...... 110 5.4.3 Conclusion ...... 112

Chapter 6. Behavioural phenotype of juvenile C57BL/6J mice exposed to DVD deficiency and PEE...... 114 6.1 Introduction ...... 114 6.2 Materials and methods ...... 116 6.2.1 Animals, housing and exposures ...... 116 6.2.2 Ultrasonic vocalizations (P7) ...... 116 6.2.3 Social interaction (P21) ...... 117 6.2.4 mRNA expression of candidate ...... 118 6.2.5 Statistical analysis ...... 119 6.3 Results ...... 120 6.3.1 Ultrasonic vocalizations (P7) ...... 120 6.3.2 Locomotion (P21) ...... 123 6.3.3 Social interaction (P21) ...... 123 6.3.4 mRNA expression of candidate genes ...... 124 6.4 Discussion ...... 128 6.4.1 DVD deficiency and PEE on ultrasonic vocalizations ...... 128 6.4.2 DVD deficiency and PEE on social interaction ...... 130 6.4.3 DVD deficiency and PEE on gene expression ...... 131 6.4.4 Conclusion ...... 133

xiv Chapter 7. General Discussion...... 135 7.1 Introduction ...... 135 7.2 Impact of DVD deficiency and PEE on neonatal outcomes ...... 136 7.3 Impact of DVD deficiency and PEE on early postnatal outcomes ...... 140 7.4 Impact of DVD deficiency and PEE on adult outcomes ...... 145 7.5 Limitations and future directions ...... 147 7.6 Conclusions ...... 148

References ...... 149

Appendices ...... 185 Appendix 1. List of primers ...... 185 Appendix 2. Group x Sex interactions in novelty-induced locomotion ...... 186 Appendix 3. Group x Sex interactions in adult hippocampal gene expression ...... 187 Appendix 4. Top molecular and cellular functions affected by DVD deficiency and PEE on gene expression ...... 188 Appendix 5. Top transcripts up and downregulated by DVD deficiency and PEE ...... 189 Appendix 6. Top molecular and cellular functions altered by DVD deficiency and PEE on protein expression ...... 190 Appendix 7. Top up and downregulated by DVD deficiency and PEE ...... 191

xv List of Figures

Figure 2.1 Maternal weight gain and drinking...... 24 Figure 2.2. Locomotion during early life (P21)...... 27 Figure 2.3 Locomotion during early adulthood (P70)...... 28 Figure 2.4. Primer validation via DNA gel electrophoresis...... 29 Figure 2.5. Expression of Vglut1 and 3 in C57BL/6J and BALB/c hippocampal tissue...... 30 Figure 2.6. Expression of Vglut2 in C57BL/6J and BALB/c hippocampal tissue...... 31 Figure 3.1. Picture of open field arena (activity monitor)...... 39 Figure 3.2. Active Place Avoidance...... 40 Figure 3.3. Locomotion during early adulthood (P70)...... 42 Figure 3.4. Spatial memory in the APA test in adult male mice...... 45 Figure 3.5. Spatial memory in the APA test in adult female mice...... 47 Figure 3.6. Spatial memory in the APA test in older adult male mice...... 49 Figure 3.7. MK-801 hyperlocomotor response in adult male mice...... 50 Figure 4.1. Maternal body weight and serum 25(OH)D levels...... 60 Figure 4.2. Blood alcohol concentration in dams at GD8...... 61 Figure 4.3. Liver ADH levels in dams at GD8...... 61 Figure 4.4 Gene expression changes in neonatal brain as a result of DVD deficiency and PEE...... 69 Figure 4.5 RNA-Sequencing results from neonatal brain tissue...... 71 Figure 4.6 IPA® network analysis of dysregulated molecules in DVD-H2O samples...... 73 Figure 4.7 IPA® network analysis of dysregulated molecules in Std-PEE samples...... 74 Figure 4.8 IPA® network analysis of dysregulated molecules in DVD-PEE samples...... 75 Figure 4.9 Protein expression changes in neonatal brain as a result of DVD deficiency and PEE...... 76 Figure 4.10 SWATH Proteomics results from neonatal brain tissue...... 78 Figure 5.1. Histological analysis of neonatal brain samples...... 101 Figure 5.2. Expression levels of affected candidate genes in P0 ...... 102 Figure 5.3. Expression levels of unaffected candidate genes in P0 cerebrum...... 103 Figure 5.4. Localization of FOXP2 and DARPP-32 in DVD-H2O mouse brain at P0...... 105 Figure 5.5. Neonatal brain anatomy...... 106 Figure 5.6. Effects of DVD deficiency and PEE on neonatal neuroanatomy...... 107 Figure 6.1. Ultravox XT software...... 117 Figure 6.2. Social Interaction...... 118 Figure 6.3. Ultrasonic vocalizations at P7...... 121 xvi Figure 6.4. Call types quantification at P7...... 122 Figure 6.5. Locomotion at P21...... 123 Figure 6.6. Social Interaction at P21...... 124 Figure 6.7. Expression levels of affected candidate genes in P21 cerebrum...... 126 Figure 7.1 Speculative trajectory of the effects of DVD deficiency and PEE in the offspring...... 135 Figure 7.2. Postulated FoxP2/Darpp-32 regulation by PEE...... 144

xvii List of Tables

Table 2.1. Developmental milestones in P21 C57BL/6J pups from DVD-deficient and PEE litters...... 25 Table 2.2. Developmental milestones in P21 BALB/c pups from DVD-deficient and PEE litters...... 26 Table 3.1. Summary of Behavioural Phenotype in Adult Offspring...... 51 Table 4.1 List of top canonical pathways altered by DVD deficiency and PEE on gene expression...... 72 Table 4.2 List of top canonical pathways altered by DVD deficiency and PEE on protein expression...... 79 Table 5.1. List of genes assessed at P0 via qRT-PCR...... 104 Table 6.1. List of candidate genes and relevant pathway assessed at P21...... 119 Table 6.2. Expression levels of unaffected candidate genes on P21 cerebrum...... 127

List of Abbreviations

Abbreviation Definition ACO1 Cytoplasmic aconitate hydratase AD Alzheimer’s disease ADH Alcohol dehydrogenase ADHD Attention deficit hyperactivity disorder APA Active place avoidance ASD spectrum disorders AVD Adult vitamin D Avy Agouti viable yellow BAC Blood alcohol concentration BCA Bicinchoninic acid Bdnf Brain-derived neurotrophic factor C4A/C4B Complement factor 4

Ca2+ Calcium CaBPS Ca2+-binding proteins cAMP Cyclic-AMP CBCL Achenbach child behaviour checklist CDR Clinical dementia rating CFlm Cleavage factor Im COTL1 Coactosin-like protein CRABP2 Cellular retinoic acid binding protein 2 Crym Crystaline mu CYP27B1 Cytochrome p450 1-hydroxylase Drd1 Dopamine 1 D2R Dopamine receptor 2 DA Dopamine DARPP-32 Dopamine- and cAMP-regulated phosphoprotein, Mr 32 kDa DENND1C Denn domain containing 1c DNA Deoxyribonucleic acid DOHaD Developmental origins of health and disease DTT Dithiothreitol DVD Developmental vitamin D EAAT1&3 Excitatory transporters 1 and 3 EPI Epinephrine ERK Extracellular signal-regulated kinase ES Early stress ETFA Electron transfer flavoprotein alpha subunit F13A1 Coagulation factor xiii a chain FA Formaldehyde FAS Foetal alcohol syndrome FASD Foetal alcohol spectrum disorders FDR False-discovery rate FGFBP3 Fibroblast growth factor binding protein 3 xix FOV Field of view FoxP2 Forkhead box protein P2 FSD1 Fibronectin type iii and spry domain containing 1 GABA Gamma-aminobutyric acid GAD65/67 Glutamate decarboxylase GD Gestational day GDNF Glial-derived neurotrophic factor GNA11 G protein subunit alpha 11 GPC2 Glypican 2 GPCRs G-protein coupled receptors H2O Water HF-D High fat diet, without vitamin D HF+D High fat diet, added vitamin D HPA Hypothalamic-pituitary-adrenal Hprt1 Hypoxanthine guanine phosphoribosyl transferase HVA Homovanilic acid IAA Idoacetamide IDA Information-dependent acquisition IGF-1 -like growth factor-1 IPA Ingenuity pathway analysis KCNQ5 Potassium voltage-gated channel subfamily q member 5 L-VGCCs L-type voltage-gated ca2+ channels LARP1 La ribonucleoprotein domain family member 1 LRRC40 Leucine-rich repeat containing protein 40 MARS Methionine-tRNA ligase MCI Mild cognitive impairment MMSE Mini mental state examination MRI Magneic resonance imaging Mrna Messenger ribonucleic acid MSNs Medium spiny MWM Morris water maze NE Norepinephrine NECAB1 N-terminal ef-hand calcium binding protein 1 NGF Nerve growth factor NMDA N-methyl-d-aspartate NR1D1 subfamily 1 group d member 1 NSCs Neural stem cells NAcc Nucleus accumbens NUDT21 Polyadenylation specificity factor subunit 5 OFC Orbitofrontal cortex PD Parkinson’s disease PEE Prenatal ethanol exposure PER3 Period circadian protein homolog 3 PFC Prefrontal cortex PGD2 Prostaglandin d2

xx PGH2 Prostaglandin H2 PP-1 Protein phosphatase-1 PPI Prepulse inhibition PSMA5 Proteasome subunit alpha type-5 PSMD13 Proteasome 26S Subunit, non-ATPase 13 PTGDS Prostaglandin d synthase qRT-PCR Quantitative real-time polymerase chain reaction RA Retinoic acid RPL23 Ribosomal protein l23 RPL7 Ribosomal protein l7 RXR SBT Short blessed test SCAMP1 Secretory carrier membrane protein 1 SCN Suprachiasmatic nuclei SCX Strong cation exchange SmithKline Beecham, Harwell, Imperial College, Royal SHIRPA London Hospital, phenotype assessment SKP1 S-phase kinase-associated protein 1 Slc17a6 Solute carrier family 17 member SND-POA Sexually dimorphic nucleus of the preoptic area Std Standard SURF4 Surfeit locus protein 4 SWATH Sequential window acquisition of all theoretical mass spectra Tfg-1 Transforming growth factor beta 1 TH Tyrosine hydroxylase TR/TE Repetition time/echo time Ubb Ubiquitin b UFM1 Ubiquitin fold modifier 1 USV Ultrasonic vocalization VDR Vglut1 Vesicular glutamate transporter 1 Vglut2 Vesicular glutamate transporter 2 VTA Ventral tegmental area WHO World health organization

xxi

1 Literature Review

Chapter 1. Literature review

1.1 Environmental risk factors for neurodevelopmental disorders

The environment in which the foetus develops is thought to be crucial in determining its immediate and future health. This is known as the Developmental Origins of Health and Disease (DOHaD) hypothesis. David Barker identified a relationship between adverse early-life environments and adverse cardiovascular disease, when he found that local authority areas within England and Wales with high infant mortality rates in 1921-1925 also showed increased mortality levels from ischaemic heart disease in 1968-1975 (Barker et al 1986). Since then, numerous longitudinal cohort studies have been established, strengthening the hypothesis that many early-life factors are associated with long-term health (Ganu et al 2012, Hanson et al 2014, Heindel et al 2017).

Environmental exposures may influence brain development at different stages, including formation and closure of the neural tube, cell differentiation and migration, formation of structures, synaptogenesis and myelination. These environmental factors are likely to act in concert with susceptibility genes, and epigenetic regulation, leading to changes in gene expression and/or to a trans generational mechanism (Lyall et al 2014). Disruptions during any of these critical stages can lead to altered neurodevelopmental trajectories that may have an effect on the child’s achievements and behaviour, potentially resulting in a diagnosable neurodevelopmental condition. Neurodevelopmental disorders are disabilities associated primarily with the functioning of the neurological system and brain, and these include attention-deficit/hyperactivity disorder (ADHD), autism, learning disabilities, (or mental retardation), conduct disorders, cerebral palsy and impairments in vision and hearing. Individuals with neurodevelopmental disorders can experience difficulties with language and speech, motor skills, behaviour, memory, learning and/or other neurological functions. Symptoms and behaviours of neurodevelopmental disabilities often change or evolve with time (for example, as a child grows older), however others are permanent.

Environmental exposures that influence neurodevelopment can include a wide range of non-genetic factors, such as medications, chemicals, physical agents, social and cultural influences as well as maternal lifestyle. These include (but are not limited to) maternal

2

nutritional status during pregnancy; use of alcohol, tobacco, or illicit drugs during pregnancy; lower socioeconomic status; preterm birth; low birth weight; the physical environment; and prenatal or childhood exposure to certain environmental contaminants (Aarnoudse-Moens et al 2009, Bhutta et al 2002, Committee on Understanding Premature et al 2006, Herrmann et al 2008, Linnet et al 2003, Perera et al 2011, Thompson et al 2007). However, environmental factors may serve as proxy markers for disease rather than causes of higher risk. For example, season is a proxy variable for various exposures, including influenza, other infections, pesticides, sunlight exposure and vitamin D levels (Lyall et al 2014). Seasonality of birth or conception has been associated with increased risk of neuropsychiatric disorders such as schizophrenia and Disorders (ASD) in several studies (Hebert et al 2010, Kolevzon et al 2006, Zerbo et al 2011). A meta-analysis found that ASD is more common among the offspring in which the first and second trimester coincided with the winter and spring months (Gardener et al 2009). Recent studies suggest that maternal diet can program offspring growth and metabolic pathways, altering lifelong susceptibility to diabetes and obesity. If maternal psychosocial experience has similar programming effects on the developing offspring, one might expect a comparable contribution to neurodevelopmental disorders, including affective disorders, schizophrenia, ASD and eating disorders. Due to their early onset, prevalence and chronicity, some of these disorders, such as depression and schizophrenia, are among the highest causes of disability worldwide according to The Lancet Global Health Series 2011 (and 2007) report. Moreover, epidemiological studies on schizophrenia have revealed that its incidence is dependent upon developmental environment, sex, ethnicity and migrant status (Bodnar et al 2007, McGrath et al 2004, Mortensen et al 1999). One such candidate environmental risk factor is developmental vitamin D (DVD) deficiency.

DVD deficiency and moderate prenatal ethanol exposure (PEE) are modifiable, environmental risk factors, which have been individually studied in the literature and have been associated with neurodevelopmental disorders, such as ASD or ADHD (Banerjee et al 2007, Cannell 2008, Eliasen et al 2010, Mukherjee et al 2011, Rojas-Mayorquín et al 2016, Vinkhuyzen et al 2017, Whitehouse et al 2013). However, no direct causality has been identified. This suggests that these complex disorders are not the result of a single exposure, but rather the combination of factors that increase the offspring’s vulnerability to eventual disease. Animal models have been a valuable tool used to test the effects of DVD deficiency and PEE, with findings in mice demonstrating that DVD deficiency has a subtle impact on brain structure, behaviour and gene expression. Some of these

3

alterations include decreased lateral ventricle volume (Harms et al 2012a), age-dependent decline in hippocampal volume (Fernandes de Abreu et al 2010), strain-dependent hyperlocomotion, increased exploration (Harms et al 2008b), altered response inhibition (Harms et al 2012b) and disrupted transcription of genes important for neural development (Harms 2011, Hawes et al 2015). Similarly, previous studies using moderate levels of PEE have shown subtle alterations in brain morphology, adult behaviour and regulation of gene expression. For example, some of the changes seen due to PEE included enlarged ventricles and hippocampus (Marjonen et al 2015), altered locomotion, learning and memory (Sanchez Vega et al 2013) and dysregulation of genes important for glutamatergic signalling (Zhang et al 2015). Therefore, the overall aim of the research for this thesis was to investigate the effect of combined DVD deficiency and PEE on brain function and behaviour in mice, to establish, for the first time, whether these exposures resulted in an increased risk of neurodevelopmental disorders.

1.2 Developmental vitamin D deficiency as a risk factor for neurodevelopmental disorders

The active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH2)D3), is a secosteroid hormone, synthesized through several intermediate stages from vitamin D (cholecalciferol). Cholecalciferol is synthesised in the skin via exposure to UBV radiation via photolysis of the prohormone 7-dehydrocholesterol (Holick 1987) or obtained from the diet, typically found in oily, deep sea fish and in lower levels in other foods such as eggs (Mattila et al 1999, Ostermeyer et al 2006). Cholecalciferol is hydroxylated in the liver to

25(OH)D3, which is then converted to the active form 1,25(OH2)D3, catalysed by the cytochrome p450 1-hydroxylase (CYP27B1). The half-life of 1,25(OH2)D3 is relatively short (approx. 15h), and its production by CYP27B1 is influenced by blood calcium and parathyroid hormone levels (Deluca 2004, Holick 2006). For this reason, 25(OH)D3 (pro- hormone) is considered a more reliable indicator of vitamin D levels.

Vitamin D is important for calcium homeostasis, bone metabolism and various other physiological functions, but also exerts potent effects on neural cells and can impact brain development and function. For example, the distribution of the vitamin D receptor (VDR) and 1 -hydroxylase (CYP27B1) has been mapped in human brain (Eyles et al 2005). In line with its receptor’s presence in developing brain tissue, vitamin D increases the

4

expression of neurotrophic factors including nerve growth factor (NGF) (Saporito et al 1994), glial-derived neurotrophic factor (GDNF) (Naveilhan et al 1996) and neurotrophic factor 3 (Neveu et al 1994), while causing down-regulation of neurotrophic factor 4 (Neveu et al 1994).

Consistent with its regulation of neurotrophic factors, there is some evidence that vitamin D is a neuroprotective agent. Vitamin D may protect against excitotoxicity induced by glutamate (Ibi et al 2001), and has been shown to have an inhibitory effect on calcium

(Ca2+) influx via down-regulation of the expression of mRNA for pore-forming subunits of

L-type voltage-gated Ca2+ channels (L-VGCCs) (Brewer et al 2001). It can also upregulate the expression of calbindin and parvalbumin in motor neurons, which are Ca2+ binding proteins that limit excitotoxicity by chelating intracellular Ca2+ (Alexianu et al 1998). The optimal level of vitamin D status, as measured by 25(OH)D3 has not yet been established.

People with circulating 25(OH)D3 < 25nmol/L are considered vitamin D deficient, and those with < 50 nmol/L are considered vitamin D insufficient (Ross 2011). Epidemiological data on women of childbearing age indicated that vitamin D deficiency was prevalent in 4% of fair-skinned women and in 42% of dark-skinned women residing throughout the United States (Nesby et al 2002), which has raised concerns regarding the impact this may have on foetal development during pregnancy. Multiple factors affect the available levels of vitamin D in the body. For example, the concentration of melanin in the skin is the major endogenous factor, as it absorbs ultraviolet B radiation, thereby inhibiting the photolysis reaction that yields vitamin D in its active form (Holick 1995). This means that dark-skinned individuals with high melanin concentrations are more prone to vitamin D deficiency than those with fair skin. In addition, exogenous factors that influence vitamin D status are mostly related to sunlight exposure (for example, season) (Wu et al 2010). Winter is associated with reduced skin exposure due to the cold as well as a reduction in sunlight intensity, which both contribute to a reduction in vitamin D synthesis (Marks 1999, Marks et al 1990, Webb et al 1988). Additionally, it is important to take into account that the decrease in sunlight intensity rises accordingly with the distance from the equator; consequently, people living at high latitudes are more likely to become vitamin D deficient during the winter months (Holick 2003).

The prevalence of maternal vitamin D deficiency is common and increasing (Looker et al 1998) and has been implicated in adverse offspring health outcomes such as ASD,

5

schizophrenia and other neurodevelopmental disorders (Ginde et al 2009, Tare et al 2006, Vinkhuyzen et al 2017). Pregnancy may be a time of particular vulnerability for sub-optimal vitamin D levels, due to dietary and lifestyle changes resulting from pregnancy. For example, a population based, case-control study, which used neonatal dried blood samples to measure the concentration of 25(OH)D3, found that neonates in the lower three quintiles (compared to those in the fourth quintile- 40.5-50.9 nmol/L) had a two-fold elevated risk of schizophrenia (McGrath et al 2010b). Another study, which used maternal mid-gestation sera examined the association between 25(OH)D3 and ASD found that mid- gestational vitamin D deficiency increased the risk of ASD by more than twofold compared to the vitamin D sufficient group (Vinkhuyzen et al 2017).

Human epidemiological studies are important to understand the prevalence of environmental risk factors, such as DVD deficiency, and the possible association with neurodevelopmental disorders. However, animal models are essential in studying how genetic and environmental risk factors impact on brain development, and how the neurobiology underlying specific behaviours under controlled conditions, while limiting confounding factors that are commonly encountered in human studies. Initially, genetic knockout models for vitamin D deficiency were highly utilised, via the homozygous ablation of the VDR gene. Behavioural effects of vitamin D deficiency showed motor impairment, altered anxiety-related behaviours, increased self-grooming and impaired prepulse inhibition (PPI) to acoustic startle (Burne et al 2005, Kalueff et al 2004). However, upon the discovery that the VDR has the ability to act independently from its ligand, it was found that a number of these abnormalities were the result of musculo-skeletal weakness due to

Ca2+/PO4 dysfunction (Minasyan et al 2009), thereby indicating that VDR ablation may not be an appropriate model of vitamin D deficiency. Consequently, the development of an alternative, more representative model of vitamin D deficiency with transient depletion of vitamin D which has a minimal confounding effect on Ca2+ or bone homeostasis was necessary.

A model of transient maternal vitamin D deficiency was initially developed in rats by feeding dams a special diet, either containing 0 (vitamin D deplete) or 1000 IU/1U/kg (standard) vitamin D for 6 weeks prior to mating with control males, throughout gestation until birth, at which point all dams were fed standard chow containing vitamin D. Using this protocol rats reach vitamin D deficiency after 6 weeks, which means that DVD-deficient rats were only deficient from conception until shortly after birth. DVD-deficient rats had

6

normal Ca2+ levels and body weight at birth and as adults (10 weeks of age), demonstrating that the temporary absence of vitamin D in this protocol did not compromise

Ca2+/bone homeostasis (Burne et al 2004b). Behavioural studies using this model of DVD deficiency have shown transient spontaneous hyperlocomotion in adult rats (Burne et al 2006). Others have found that DVD deficiency is associated with subtle changes in learning and memory functions as adults. In particular, a significantly impaired latent inhibition was found, however both memory acquisition and retrieval were unaffected (Becker et al 2005). Furthermore, rat pups born from vitamin D depleted mothers had an increased number of mitotic cells and fewer apoptotic cells in brain tissue at birth, which consequently suggests that low 25(OH)D3 concentrations can cause transcriptional deregulation in the brain. Moreover, with regards to brain structure, pups born from vitamin D depleted mothers had enlarged lateral ventricles and extended but proportionally thinner cortices. The increased lateral ventricular volume and alterations in gene and protein expression persisted into adulthood, which was associated with altered behaviour in response to psychomimetic drugs (Kesby et al 2006, Kesby et al 2012). Morphological changes in the brain, including thinning of and ventricle overgrowth, have been associated with psychiatric disorders such as schizophrenia.

A model of DVD deficiency was also established in two mouse strains, C57Bl/6J and 129/SyJ. After a comprehensive behavioural screen, it was found that 129/SvJ DVD- deficient mice exhibited spontaneous hyperlocomotion in the open field, and 129/SvJ and C57BL/6J DVD deficient mice were hyper-explorative on the hole-board test (Harms 2011). There was no effect of maternal diet on parameters assessed by the SmithKline Beecham, Harwell, Imperial College, Royal London Hospital, phenotype assessment (SHIRPA) primary screen, a procedure is used for the standardised assessment of mouse phenotype, or on tests of sensorimotor gating, social behaviour, anxiety or depression. Another DVD deficiency study using BALB/c mice focused on foetal development and found structural and gene expression changes in the foetus (Hawes et al 2015). For example, stereological analysis showed a reduction in foetal crown-rump length and head size, as well as in lateral ventricle volume of DVD deficient samples at E17.5. In terms of gene expression, DVD deficiency was associated with changes in Forkhead box protein P2 (FoxP2) and tyrosine hydroxylase (Th), which are genes related to neuronal survival, speech and language development and dopamine synthesis, thereby providing further evidence to suggest that vitamin D plays part in mouse neurodevelopment (Hawes et al 2015). However, the behavioural phenotype of these mice was not characterized in this

7

study, although it would be valuable to study the long-term effects of the alterations found during foetal development.

Some behavioural findings, such as hyperlocomotion, are consistent across rats and mice, but there were a number of behavioural differences reported in DVD-deficient mice that were not observed in rats, such as increased exploration (found in mice (Harms et al 2008a), not in rats (Becker et al 2005, Burne et al 2004b)), while behavioural response to psychomimetics was altered in rats (Kesby et al 2006) but not displayed in C57BL/6J or 129/Svj mice (Harms et al 2012a). Additionally, findings were also dependant on the background mouse strain, as there were discrepancies between C57BL/6J and 129/Svj. Although the behaviour of DVD-deficient BALB/c mice has not been characterized, adult vitamin D deficiency studies have shown that the behavioural profile of BALB/c mice is significantly different to that of C57BL/6J, showing altered behaviour in anxiety-related tasks, as well as altered responses to heat, shock and sound (Groves et al 2013).

Taken together, temporary depletion of vitamin D during gestation in mice results in subtle alterations in the offspring, suggesting that it is a plausible risk factor for neurodevelopmental disorders. However, no direct causal relationship between vitamin D and brain disorders has been found. It has now been established that the majority of neurodevelopmental disorders, including ASD, ADHD and schizophrenia are a result of a combination of genetic and/or environmental factors. Therefore, it is possible that sub- optimal vitamin D levels reduce the neuroprotective capacity leading to increased vulnerability to insults from secondary exposures, such as detrimental environmental exposures, injury or disease (Balden et al 2012, Suzuki et al 2013). The summation of these factors may result in greater risk of neurodevelopmental disorders.

There is preclinical evidence that vitamin D deficiency can exacerbate second hit exposures. For example, a study using a model of stroke in rats showed that vitamin D- deficient rats had a larger infarct volume in comparison to controls, which then correlated with greater post-stroke impairments in sensorimotor behavioural tests (Balden et al 2012). Molecular analyses revealed that compared to controls, vitamin D-deficient rats had significantly lower levels of Insulin-like Growth Factor-1 (IGF-1) in plasma, brain and liver. IGF-1 is a neuroprotectant normally elevated post-injury to rescue the tissue. Therefore, it is possible that a reduction of IGF-1 occurred as a result of vitamin D deficiency, which then contributed to the greater infarct volume observed in vitamin D-deficient rats (Balden et al 2012). Another study focusing on Parkinson’s Disease (PD) provided further evidence 8

for the neuroprotective properties of vitamin D. The impact of vitamin D supplementation (1200 IU daily for one year) was examined on a number of PD related outcomes. Patients that belonged to the Placebo group showed a stable worsening of PD outcomes, while those in the vitamin D supplement group presented no change in PD outcomes throughout the year of treatment, which suggests that low levels of vitamin D exacerbate the progression of disease. It has also been suggested that low vitamin D can aggravate the vulnerability to psychosocial stressors (McGrath et al 2010a). Jiang et al (2013) documented an up-regulation of both VDR and 1,25(OH)2D in the hippocampus of rats as a result of chronic mild stress. This suggests that vitamin D signalling may be essential for compensatory mechanisms that take place to protect the brain from insults such as stress, and that the combination of both vitamin D deficiency and a secondary exposure may result in greater disease susceptibility.

1.3 Secondary insults to DVD deficiency

A second hit exposure may occur at various time points: prenatally (concurrently with maternal vitamin D deficiency), during early postnatal life or in adulthood. Some prenatal exposures that are commonly observed in the population are exposure to toxins, use of tobacco, illicit drugs and alcohol. In this review, I will focus on prenatal ethanol exposure (PEE) as a secondary hit exposure to DVD deficiency. Australian statistics have reported that alcohol consumption in women of childbearing age to be as high as 85%. Moreover, with over 50% of pregnancies unplanned (Giglia et al 2007), this may allow at least a four- week gap during which pregnancy might be unknown and alcohol consumption may occur.

The deleterious effects of prenatal alcohol consumption were first established in 1973 and are referred to as Foetal Alcohol Syndrome (FAS) (Jones et al 1973). FAS is characterized by three main clinical features, including growth restriction, craniofacial abnormalities in addition to structural and/or functional deficits in the brain (Abel 1984, Sokol et al 2003). It is the most severe form of alcohol related disorders within the non-diagnostic umbrella term foetal alcohol spectrum disorders (FASD). Although the severity can vary among individuals, it is mostly associated with the level of exposure, in which heavy drinking patterns are more likely to cause FAS. Moderate and light drinking are commonly responsible for milder forms of FASD, which lack the presence of dysmorphic features typical of FAS but exhibit neurobehavioural and cognitive impairment (Abel et al 1998). For

9

example, several studies have reported FASD children to be hyperactive, irritable and experience difficulties in tasks of vigilance, reaction time and information processing. Life- long impairments can include impulsivity, hyperactivity, social ineptness, poor judgement and learning disabilities (Brown et al 1991, Coles et al 2011, Mattson et al 2011, Nanson et al 1990).

FAS is the least common effect of prenatal alcohol exposure among the FASD umbrella, with a prevalence of 5% in children of typically alcoholic mothers (Abel et al 1998, Kaminen-Ahola et al 2010a). A major focus of prenatal alcohol research has been on levels of alcohol within the moderate to high range. Fewer studies have investigated the impact of relatively low concentrations of alcohol exposure in utero on behaviour and cognitive performance in the offspring, with studies reporting conflicting findings. A large longitudinal study in pregnant women revealed that mothers who consumed light levels of alcohol (2-6 standard drinks per week) in the first three months of pregnancy had children with significantly lower total and internalizing behaviour scores over fourteen years, representing more positive behaviour than non-drinkers at three months’ gestation (Robinson et al 2010). This study also revealed that children of light to moderate drinkers (2-10 standard drinks per week) were at a clinically meaningful lower risk of total, internalising and externalising behavioural problems than the children of women who did not drink. Although these results support other studies with similar findings, which suggest that low levels of alcohol exposure were not associated with developmental risks (Kelly et al 2009), they are in opposition to others, which report that even low levels of alcohol were adversely related with child behaviour (Sayal et al 2007); (Sood et al 2001). A prospective, population-based study found that consumption of >1 drink per week during the first trimester of pregnancy was independently associated with clinically significant mental health problems in girls at 47 months, and persisted at 81 months (Sayal et al 2007). Another longitudinal study monitored maternal alcohol and drug use during pregnancy, and followed up with families six years later. Using the Achenbach Child Behaviour Checklist (CBCL) to assess child behaviour, results showed that children with any prenatal alcohol exposure were more likely to have higher CBCL scores on externalizing (aggressive and delinquent) and internalizing (anxious/depressed and withdrawn) syndrome scales as well as on the total problem score. Furthermore, there were significant differences found between no and low-exposure groups for aggressive and externalizing behaviours at age 6 to 7, which persisted after controlling for other factors associated with

10

adverse behavioural outcomes, thereby suggesting that the adverse effects of prenatal alcohol exposure are evident even at low levels of exposure (Sood et al 2001).

Rodents have been widely used to investigate the effect of prenatal ethanol exposure on brain function and behaviour (Kelly et al 2009, Wilson et al 2011). However, there is no standard FASD rodent model because researchers alter experimental parameters such as dose, pattern and timing of exposure and route of administration, which influence outcomes and mirrors the diverse manifestations of FASD in humans. C57BL/6J mice exhibit a propensity to voluntarily consume alcohol (Kleiber et al 2011) which has a practical advantage in that it minimizes confounding effects of maternal stress caused by ethanol injections. A previously established mouse model utilised ad libitum ethanol exposure (moderate levels of 10% v/v) during the first eight days of gestation (GD 0-8), which is equivalent to the first 3–4 weeks of a human pregnancy, thereby mimicking the period of time during which mothers are unaware of pregnancy (Kaminen-Ahola et al 2010a, Kaminen-Ahola et al 2010b). Using this procedure, adult coat colour changes were detected in C57Bl/6 mice carrying the epigenetically regulated allele, Agouti viable yellow (Avy). Given that adult coat colour is linked to the Avy allele, changes in this phenotype show that moderate prenatal alcohol exposure (GD 0–8) could alter Avy expression and DNA methylation. This study showed, for the first time, that moderate prenatal alcohol exposure could affect the adult phenotype by modifying the epigenotype of the early .

With regards to behaviour, social and cognitive domains have been studied across various models of ethanol exposure. As previously mentioned, experimental design (dose, timing and mode of exposure) greatly influences on outcome measures, therefore, mixed findings have been reported. Communication in mice is often assessed via ultrasonic vocalizations (USVs) emitted by pups when separated from the dam and litter (Scattoni et al 2009b). A study in which dams were exposed to ethanol for the first and second trimester reported a reduction in USVs in PEE juvenile offspring (Wellmann et al 2015), while a model of chronic exposure (GD1-P10) showed no effect of ethanol. Furthermore, in the sociability domain, some have reported reduced play/fight encounters, shorter gap-crossings (Wellmann et al 2015), impaired prosocial interaction and increased avoidance (Hellemans et al 2010) and increased wrestling behaviour in ethanol-exposed adult males, while little to no effect has been found in females. Moreover, there are little to no reports on these

11

behaviours at juvenile stages, during which neurodevelopmental disorders as ASD usually become prominent in children.

Given that learning and memory deficits have been a prominent phenotype in children with FAS, this domain has been heavily studied in rodent models utilising several methods. High levels of exposure have found deficits in the Morris Water Maze (MWM) in rats (Savage et al 2002) (Mattson et al 1999), and similarly, chronic ethanol exposure in mice has been shown to induce deficits in the Barnes Maze test (Kleiber et al 2011); nevertheless, moderate levels of exposure have been reported to have either no effect in rats (Cullen et al 2013), while another study in mice showed an enhanced performance in ethanol-exposed males in the MWM (Sanchez Vega et al 2013). Based on its crucial role in learning and memory, the hippocampus has been the focus of numerous prenatal alcohol exposure studies. Human data has shown a reduction in hippocampal volume and Magneic Resonance Imaging (MRI)-behavioural evaluations have suggested links between alterations in hippocampal volume and memory deficits (Coles et al 2011, Willoughby et al 2008). Likewise, decreased hippocampal volume has been reported in rats (Livy et al 2003, Parnell et al 2009) along with alterations in hippocampal-dependent learning and memory (Wozniak et al 2004, Zink et al 2011).

At a molecular level, studies using high dose ethanol exposures have encountered alterations in glutamatergic signalling as well as in synaptic plasticity in hippocampal tissue, showing dysregulation of vesicular glutamate transporter 1 (Vglut1), complexin 1, various N-methyl-d-aspartate (NMDA) receptor subunits and excitatory amino acid transporters 1 and 3 (EAAT1 and 3) (Naassila et al 2002, Zink et al 2009, Zink et al 2011). Moreover, a recent model using moderate levels of exposure early in gestation revealed a disruption in the developmental silencing of solute carrier family 17 member (Slc17a6), which encodes for vesicular glutamate transporter 2 (Vglut2) in the adult hippocampus, suggesting a long-term effect on glutamatergic signalling. Additionally, epigenetic analyses have shown complex patterns of DNA methylation and post-translational histone modifications at the promoter and an upstream region of Slc17a6, including dynamic epigenetic marks that are altered by transcriptional activity as well as fixed DNA methylation mark that possibly confers epigenetic memory of prenatal ethanol exposure.

PEE has also been found to affect other systems, including the dopaminergic system (Druse et al 1990). PEE has been associated with a reduction in

12

spontaneous activity in dopamine (DA) neurons, decreased DA uptake and receptor binding sites, decreased DA metabolite homovanilic acid (HVA) in DA neurons and decreased D1 receptors in the hypothalamus and striatum (Cooper et al 1988, Detering et al 1980, Druse et al 1990, Rathbun et al 1985, Shen et al 1999, Zhou et al 2003), as well reduced dendritic growth in DA neurons (Shetty et al 1993) and changes in DA receptor function (Shen et al 1995), which highlights the potent extent to which ethanol can affect neurodevelopment.

Taking into account the prevalence and biological plausibility of both DVD deficiency and moderate PEE, as well as evidence showing both of these factors have subtle effects on offspring behaviour and can impact glutamatergic and dopaminergic signalling, an investigation of the combined effects of these exposures in the offspring is warranted.

1.4 Aims & outline of thesis

The first aim for the research presented in this thesis was to develop a two-hit model of DVD deficiency and PEE in mice. It was hypothesized that BALB/c mice would be more vulnerable to the effects of these exposures compared to C57BL/6J mice. This was examined in Chapter 2.

The second aim was to determine the effects of DVD deficiency and PEE on neonatal gene and protein expression, as well as on brain structure. It was hypothesized that offspring from the two-hit exposure group would show further alterations when compared to those exposed to DVD deficiency or PEE alone. A broad screening approach was used to verify the effects of DVD deficiency and PEE on transcript and protein expression and this was examined in Chapter 4. This was followed by a more focused analysis, where the expression of specific candidate genes important for neurodevelopment was evaluated in Chapter 5. The effects of DVD deficiency and PEE on brain morphology was also assessed via MRI and histological analysis in Chapter 5.

The third aim of this thesis was to study the effects of DVD deficiency and PEE on behaviour. It was hypothesized that the absence of vitamin D during development would exacerbate previously described effects of PEE on adult behaviours, including altered learning and memory and response to psychomimetic drugs. These data are presented in Chapter 3. The effects of DVD deficiency and PEE on juvenile behaviour were examined 13

in Chapter 6, focusing on behaviours known to be disrupted in early-onset neurodevelopmental disorders, such as ASD, including communication, locomotion and sociability.

14

Establishing a two-hit model of DVD deficiency and PEE in 2 C57BL/6J and BALB/c mice

Chapter 2. Establishing a two-hit model of DVD deficiency and PEE in C57BL/6J and BALB/c mice

2.1 Introduction

Epidemiological studies indicate that vitamin D deficiency in Australia is prevalent in women of childbearing age (Bodnar et al 2007, Daly et al 2012). It is now known that DVD deficiency has a long-lasting impact on both behavioural and neurochemical characteristics in the offspring (Burne et al 2004a, Eyles et al 2007, Fernandes de Abreu et al 2010). PEE is also a plausible risk factor for long-lasting consequences in offspring because of elevated alcohol consumption rates in women of childbearing age and the large proportion of unplanned pregnancies (approx. 50%) (Colvin et al 2007). Furthermore, there is an increased likelihood that these two common exposures take place simultaneously, however, there is a gap in the literature focusing on their combined risk, therefore, it is important to address the effects the combined effects of DVD deficiency and PEE on development and later in adulthood.

C57BL/6J mice are a widely used strain with a well-known genetic background. However, there are considerable behavioural differences between mouse strains, for example Crawley et al (1997), have shown that other strains, such as BALB/c are more sensitive/vulnerable to subtle exposures, making it easier to detect altered phenotypes than in C57BL/6J mice (considered a resilient strain). A small number of studies have compared the effects of PEE in C57BL/6J to other strains. One study compared C57BL/6J to BALB/c mice and focused on the effects of PEE on the development of the corpus callosum, and found a reduction in the area of the corpus callosum and the anterior commissure in PEE BALB/c mice, while the same parameters were not altered in PEE C57BL/6J mice (Wainwright et al 1985). In a different study looking at the effects of PEE on adult behaviour, where C57BL/6J mice were compared to C3H/He mice, male C3H/He PEE offspring showed impaired scent marking behaviour compared to controls, while C57BL/6J did not show changes in scent marking, suggesting they were more resilient to the effects of PEE (Hale et al 1992). Nevertheless, another PEE study comparing C57BL/6J mice to DBA/2J mice found different gene expression patterns between strains, but only C57BL/6J mice showed alterations in gene expression (genes relevant to mRNA

16 splicing, transcription and translation) as a result of ethanol exposure (Downing et al 2012).

With respect to vitamin D deficiency, only one study of DVD deficiency has directly compared two inbred mouse strains (C57BL/6J and 129/SVJ mice). Neonatal DVD- deficient mice of both strains had increased hippocampal volumes compared to control mice. Increased exploration was also found in adult DVD-deficient offspring of both strains (Harms et al 2008a). However, hyperlocomotion, the most replicated behavioural phenotype in rat DVD deficiency models (Burne et al 2006), was only found in 129/SVJ mice. By contrast, hyperlocomotion was found in C57BL/6J and BALB/c strains using a mouse model of Adult vitamin D (AVD) deficiency, whereas BALB/c mice also showed altered anxiety-like behaviours, while C57BL/6J did not. Furthermore, neurochemistry analysis reported marked differences between strains upon AVD deficiency, in which C57BL/6J mice had increased dopamine and 5-hydroxytryptamine turnover, while the BALB/c strain showed decreases in levels of glutamate and glutamine and increased levels of gamma-aminobutyric acid (GABA) and glycine. However, both inbred mouse strains showed a small but significant decrease in the level of an enzyme involved in GABA synthesis, glutamate decarboxylase (GAD65/67) (Groves et al 2013). Overall, these studies demonstrate the importance of background strain, and in the case of moderate environmental exposures as low-dose ethanol exposure and vitamin D deficiency, suggest that perhaps C57BL/6J is not the optimal strain, and a more sensitive strain may be more appropriate. Therefore, the first aim of this experiment was to compare the effects of DVD deficiency and PEE on C57BL/6J to BALB/c mice, to then select one strain for future experiments.

A number of PEE studies in rodents have reported abnormal growth and development as a result of the exposure to ethanol. However, the dose and duration of exposure may play an important role in the phenotype of the offspring (Mantha et al 2013, Tran et al 2000). There are no previous DVD deficiency studies that have examined postnatal development in mice. Therefore, the second aim of this experiment was to test whether the combination of PEE and DVD deficiency had a significant effect on the achievement of early developmental milestones and if there were any long-term changes observed during adulthood. Locomotion is a simple but sensitive parameter that would provide an initial insight as to whether the adult phenotype is altered in DVD-PEE animals. Both PEE and DVD deficiency studies have individually shown alterations in locomotion, though results

17 are not consistent across studies (hyper/hypo) and vary with strain, dosage and housing environment (Abate et al 2017, Burne et al 2004a, Harms et al 2008a, Sanchez Vega et al 2013).

The precise molecular mechanisms by which either DVD deficiency or PEE influences long-term behaviour in the offspring are not known. The concept of a two-hit model has been used to investigate whether there is an additive effect seen whereby one exposure may act in concert with the other and have a synergistic effect (Balden et al 2012, Jiang et al 2013, McGrath et al 2010a, Suzuki et al 2013). At a molecular level, studies using high dose ethanol exposures have encountered alterations in glutamatergic signalling, and changes in synaptic plasticity in hippocampal tissue have been implicated, through dysregulation of VGLUT1, complexin 1, various NMDA receptor subunits and EAAT1 and 3 (Naassila et al 2002, Zink et al 2009, Zink et al 2011). Moreover, a recent model using moderate levels of exposure early in gestation revealed a disruption in the developmental silencing of Slc17a6, which encodes for VGLUT2 in the adult hippocampus, suggesting a long-term effect on glutamatergic signalling (Zhang et al 2015). Additionally, epigenetic analyses have shown complex patterns of DNA methylation and post-translational histone modifications at the promoter and an upstream region of Slc17a6, including dynamic epigenetic marks that are altered by transcriptional activity as well as fixed DNA methylation mark that possibly confers epigenetic memory of PEE (Zhang 2013).

There are limited data examining glutamate regulation in DVD mice, although studies in rats have shown changes in glutamate and dopamine signalling from birth. For example, there was an increase of dopamine in the basal ganglia, increase in noradrenaline levels and a decrease of glutamate and GABA in the hippocampus of neonatal DVD-deficient rats (Kesby et al 2017), suggesting that DVD deficiency may play a part in the regulation of both the glutamate-glutamine cycle and dopamine signalling.

Therefore, the last aim of this experiment was to study the effects of DVD deficiency and PEE on glutamatergic regulation, starting with its vesicular transporters (Vglut1-3). Vglut1 expression is upregulated around birth and peaks at 2 week of age, whereas Vglut2 expression is widely distributed during embryogenesis, peaks at around birth, and subsequently becomes downregulated in cells in which Vglut1 is progressively upregulated (Fremeau et al 2004). Genetically modified mice with either Vglut1 or Vglut2 deletion lead to a loss of glutamatergic transmission from the affected neurons. Not

18 surprisingly, the deletion mutants suffer from severe pathological symptoms (Fremeau et al 2004, Wojcik et al 2004) or die at birth (Moechars et al 2006, Wallen-Mackenzie et al 2009). Moreover, a conditional knockout- Vglut2f/f; DAT-Cre cKO mouse line displayed no phenotypic alterations compared to wild-type litter mates, although mutant subjects had an altered behavioural phenotype, showing hyperactivity, reduced anxiety-related behaviour and increased risk-taking behaviour (Wallen-Mackenzie et al 2009).

Therefore, the key outcome measures of interest in the current experiment were developmental milestone assessment (P21) locomotion (P21 and P70) and adult hippocampal expression of Vglut1-3 in the offspring of vitamin D deficient and ethanol exposed dams of both C57BL/6J and BALB/c mice.

2.2 Material and methods

2.2.1 Animals and housing

Twenty adult male and 40 four-week old female mice of each strain (C57BL/6J and BALB/c) were purchased (ARC, Perth, WA) and housed in individually ventilated OptiMice cages (Animal Care Systems, CO, USA) at 21°C, 40–60% humidity and 12 h light dark cycle (lights on 0700 h) with ad libitum access to pelleted food (Specialty Feeds, WA) and water. The mice were habituated to the QBI animal house for 4–5 days and the females were divided across four breeding waves of 20 females each, two waves per strain, and separate waves of mice arrived every fortnight. The same sires were used throughout both waves of each strain to reduce variability from different sires. Dams from each strain were randomly allocated to one of four experimental groups: 1) Standard diet and water (Std-

H2O), 2) Standard diet and ethanol (Std-PEE), 3) vitamin D deplete diet and water (DVD-

H2O) or 4) vitamin D deplete diet and ethanol (DVD-PEE). All experimental work was performed with approval from the University of Queensland Animal Ethics Committee (QBI/461/11/NHMRC; QBI/461/14/NHMRC), under the guidelines of the National Health and Medical Research Council of Australia.

19

2.2.2 Vitamin D deficiency

Prenatal vitamin D deficiency was achieved using a previously established method by feeding female mice from four weeks of age a diet free of vitamin D (vitamin D Deficient AIN93G Rodent diet, Specialty Feeds, WA, Australia), and by housing all mice under incandescent lighting free of UVB radiation as described in (Harms et al 2008a). Standard diet (control) animals were housed in the same conditions as vitamin D deficient animals, but were given a standard vitamin D-containing diet (Standard AIN93G Rodent diet, Specialty Feeds, WA, Australia). At 10 weeks of age, female mice were caged with a single vitamin D replete male in the afternoon and checked each morning between 0830 and 0900 h for the presence of a vaginal plug, which indicated that mating had taken place. The day of plug detection was considered gestational day 0 (GD 0), at which time the female was removed from the cage, housed individually and randomly allocated to an exposure group (see maternal ethanol exposure below). Dams remained on these respective diets throughout gestation. All dams were placed on the control diet (containing vitamin D) within 12 h of giving birth. Offspring were weaned 28 days after birth, and were provided with water and standard vitamin D containing mouse pellets (Feeder and Grower diet, Specialty Feeds, WA, Australia) for the remainder of the experiment.

2.2.3 Maternal ethanol exposure

Prenatal ethanol exposure was carried out as previously described (Kaminen-Ahola et al 2010a, Kaminen-Ahola et al 2010b). Briefly, immediately after plug detection, females from both vitamin D deplete and standard diets were randomly assigned to the ethanol group and they had access to one bottle containing 10% ethanol for 8 days, which is equivalent to the first 3–4 weeks of a human pregnancy, thereby mimicking the period of time during which mothers are unaware of pregnancy. The remaining mice comprised the water group, which had access to a bottle containing tap water. All animals received ad libitum access to the drinking bottles and food (either vitamin D depleted or standard pellets). Every 24 h the contents of the water bottles were replaced with fresh solution and consumption (ml) measured. On the last day of exposure, GD8, the bottle containing ethanol was replaced with a bottle containing water and body weight was measured. All dams were subjected to only one cycle of ethanol exposure, and all other environmental factors (e.g. cage type, environmental enrichment) were kept consistent between groups.

20

2.2.4 Behaviour

2.2.4.1 Early developmental measures

Developmental differences between exposure groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) were measured at P21 in mice from all litters. Measures were assessed at P7 in preliminary data collection, however, cannibalism was a significant issue, therefore, further testing proceeded with P21 assessment only in order to maximise litter numbers for further experiments. The parameters measured were body weight, body length, ear folding and eye opening (scale 0-2) and righting reflex (scale 0-1) (adapted from Fox (1965)). In addition, the number of steps made in 30 s was recorded as a measure of locomotion.

2.2.4.2 Adult locomotion

C57BL/6J and BALB/c mice from all four experimental groups were assessed for locomotor activity at P70. Each mouse was placed in a clear open field (27.5 x 27.5 x 30cm, Med Associates Inc, USA) within a sound attenuated chamber and activity levels were recorded for 30 min. The light level was set at 18 lux. As a measure of spontaneous activity, distance travelled was calculated using activity monitor tracking software based on beam breaks from three 16 beam infrared arrays and was sampled in 1 min time bins (Stanford 2007, Walsh et al 1976).

2.2.5 Molecular

2.2.5.1 Tissue collection

Following behavioural analysis, brain tissue was collected from a total of 205 offspring at P70-77 (24h after locomotion test). Each mouse was lightly anaesthetized with isofluorane (for < 20 s) until motor control was lost and euthanized rapidly by cervical dislocation and then decapitation. The brain was then removed from the skull and the entire hippocampus was then dissected from the remaining posterior block of brain tissue. All tissues were snap-frozen in liquid nitrogen within 5 min of death and then stored at -80˚C.

21

2.2.5.2 Primer validation

Prior to performing quantitative real-time polymerase chain reaction (qRT-PCR) analysis on experimental samples, primers were tested for dimer formations and housekeeping genes were selected (See Appendix 1 for primer sequences). DNA gel electrophoresis was carried out for a more accurate visualization of results. Briefly, a 2% agarose gel with added SYBR safe stain was prepared and 20 µl of samples were loaded in each well (samples were prepared by adding 25µl of qRT-PCR product + 5µl 1xDNA sample dye) with a DNA reference ladder in the middle lane; the gel was run in 0.5x TBE buffer at 75V for 55min and then visualized under UV light.

2.2.5.3 qRT-PCR

A total of 91 hippocampal tissue samples were selected for gene expression analysis (n= 7-12/Group/Sex). RNA (1ug) was reverse transcribed in a 20 µl reaction using 0.5μg/ml Oligo(dT), 10mM dNTP mix, 10x RT buffer, 0.1M DTT, 40 units/μl of RNaseOUT and 200 units/μl SuperScript III Reverse Transcriptase. RT reactions were diluted 10 times and 3 µl was used in the qRT-PCR with 1 x SYBR green MasterMix and 1 µM of forward/reverse primers. Cycling parameters included 50°C for 5 min, followed by 40 cycles of denaturation at 95°C for 15 s and annealing and extension at 60°C for 1 min. Every sample was loaded in triplicate in each qRT-PCR plate and plates were performed in triplicate to confirm the validity of the results. Hypoxanthine guanine phosphoribosyl transferase (Hprt1) was used as housekeeper. Analysis of qRT-PCRs utilised the comparative Ct method (Schmittgen et al 2008). Primer sequences for qRT-PCR analysis of Vglut1-3 were obtained from previous literature (Cheng et al 2011); Hprt1 was chosen as housekeeping gene, based on both the literature and their consistent and high level of transcription in the hippocampal samples previously assayed by microarray. Its expression levels were assessed across all samples using the Ct values from the qPCR reaction and performing a one-way ANOVA to confirm there were no significant changes in expression across experimental groups (Diet: F1,89=0.00, p=0.99; Exposure: Diet: F1,89=0.62, p=0.43;

DietxExposure: F1,89=0.99, p=0.32). The transcriptional levels of Vglut1-3 were analysed by qRT-PCR in adult hippocampal samples of both sexes (Waves 1, 2 and 3).

22

2.2.6 Statistical analysis

Results were analysed for statistical significance using the SPSS statistics software package (ver. 24, SPSS Inc., Chicago, Illinois). There were no significant Group x Sex interactions, therefore, data were presented pooled for sex (Appendix 2 & 3). Data were analysed using ANOVA and, where appropriate, repeated measures were used. Student’s t-test was used to assess the difference between two groups. Associations between behavioural results and gene expression levels were performed using non-linear regression. A p value of <0.05 was considered to be statistically significant.

2.3 Results

2.3.1 Breeding outcomes

There was no significant effect of maternal Diet on pregnancy rate, litter size or dam and pup mortality in mice. The average daily consumption of 10% ethanol during GD0–8 was 2.5±0.2 (SEM) ml/mouse/day (or 12.5 g ethanol/kg body weight/day) and was not different between strains. It has been shown that female mice voluntarily consuming 10% ethanol at 14 g ethanol/kg body weight/day produces average peak blood alcohol levels of ca. 120 mg/dl (Allan et al 2003). There was no significant effect of Diet (F1,88=1.7 p= 0.2; Fig. 2.1

A) or Exposure (F1,88=3.6 p= 0.06; Fig. 2.1 A) in maternal weight gain across the ethanol exposure days (GD0-8). Average drinking volumes (measured daily GD0-8) were not affected by Exposure (F1,88=2.8, p=0.1; Fig. 2.1 B). Inspection of Fig. 2.1B suggests that DVD-deficient dams drank more than Std-diet mice. However, the main effect of Diet did not reach significance (F1,88=3.6, p=0.06; Fig. 2.1 B).

23

Maternal Weight Gain (0.5-8.5dpc) Maternal Drinking Volume (0.5-8.5dpc) A B 2.5 4 Std-H O

) 2 l

2.0 m (

3 DVD-H2O

e

)

g

m (

1.5 Std-PEE

u

t

l h

o 2

g V

i DVD-PEE

e

1.0 e

g

W a r 1

0.5 e

v A 0.0 0

Figure 2.1 Maternal weight gain and drinking. (A) There was no significant effect of Group in weight gain between the first 8 days of gestation, which coincide with exposure to ethanol or water. (B) There was no effect of Group in the average drinking volumes across the first 8 days of gestation. Data presented as mean  SEM.

2.3.2 Developmental milestone assessment at P21

The postnatal development of 205 offspring from all Groups was monitored at P21. Monitoring initially intended to take place at P7 and P14, however this was later was limited to P21 to avoid cannibalism induced by handling. There was no significant effect of Group (either Diet or Exposure) on body weight and length, ear folding, eye opening, and righting reflex (Tables 2.1 and 2.2). There was a significant main effect of Strain on locomotion, measured as the number of steps in 30 s, whereby C57BL/6J mice showed increased locomotion at P21 in comparison to BALB/c mice. Likewise, a main effect of Diet

(F1,208=74.013, p<0.001; Fig 2.2) and Exposure (F1,208=123.653, p<0.001; Fig. 2.2) was found on locomotion, as well as a significant interaction of Diet x Exposure, whereby DVD- PEE mice showed a significant increase in locomotion at P21 compared to controls (Std-

H2O) (F1,208=11.853, p<0.001; Fig. 2.2).

24

Table 2.1. Developmental milestones in P21 C57BL/6J pups from DVD-deficient and PEE litters.

C57BL/6J

Std-H2O (n=20) Std-PEE (n=7) DVD-H2O (n=24) DVD-PEE (n=27)

SE Mean SEM Mean Mean SEM Mean SEM M Weight (g) 7.68 0.25 6.93 0.46 7.06 0.17 7.11 0.14 Length-full (cm) 11.44 0.17 10.24 0.25 10.96 0.12 10.98 0.1 Eyes.opening (0-2) 2 0 2 0 2 0 2 0 Ear folding (0-2) 2 0 2 0 2 0 2 0 Righting reflex (0-1) 1 0 1 0 1 0 1 0 Locomotion.d21 42.15 1.56 48.29 2.49 49.54 1.19 58.74 0.96 (steps/30s)

25

Table 2.2. Developmental milestones in P21 BALB/c pups from DVD-deficient and PEE litters.

BALB/c

Std-H2O (n=36) Std-PEE (n=20) DVD-H2O (n=30) DVD-PEE (n=44)

SE Mean SEM Mean SEM Mean Mean SEM M Weight (g) 9.69 0.24 10.03 0.19 9.67 0.4 9.31 0.17 Length-full (cm) 12.48 0.08 12.53 0.13 12.61 0.12 12.48 0.08 Eyes.opening (0-2) 2 0 2 0 2 0 2 0 Ear folding (0-2) 2 0 2 0 2 0 2 0 Righting reflex (0-1) 1 0 1 0 1 0 1 0 Locomotion.d21 26.19 0.88 42.55 1.26 36.27 1.17 49 1.15 (steps/30s)

26

Main effect of Diet: (F1,208=74.013, p<0.001) Main effect of Exposure: (F1,208=123.653, p<0.001) Main effect of Diet x Exposure: (F1,208=11.853, p<0.001)

C57BL6/J P21 BALB/c P21 A B 60 60

Std-H2O

DVD-H O n

n 2 e

40 e 40 k

k Std-PEE

a

a

t

t

DVD-PEE

s

s

p

p

e

e t

20 t 20

S S

0 0

Figure 2.2. Locomotion during early life (P21). (A) Locomotion was measured in C57BL/6J mice at P21 as numbers of steps taken in 30s. Exposure to DVD deficiency alone, as well as PEE increased activity levels; exposure to both DVD deficiency and PEE further increased locomotion. (B) Locomotion in BALB/c mice at P21. PEE increased locomotion; similar effects were seen after DVD deficiency. Increased hyperlocomotion was noted as a result of the combination of DVD-PEE. Data presented as mean  SEM.

2.3.3 Adult locomotion (P70)

Novelty-induced locomotion from all animals was evaluated in an Activity Monitor for 30min. There was a significant effect of Strain, whereby C57BL/6J mice were more active than BALB/c. Analysis split by Strain showed that there was a main effect of Diet (F1,74=

8.3 p=0.005; Fig. 2.3 A) and of Exposure (F1,74= 7.2 p=0.009; Fig. 2.3 A), but not an interaction of Diet x Exposure (F1,74= 2.01 p=0.16; Fig. 2.3 A) in C57BL/6J mice. There were no significant effects of Diet (F1,129=0.53 p=0.47; Fig. 2.3 B), Exposure (F1,129=1.2 p=0.27; Fig. 2.3 B) or an interaction of Diet x Exposure (F1,129=2.2 p=0.14; Fig. 2.3 B) in BALB/c mice. The exacerbated hyperlocomotive effect of Diet and Exposure seen at P21 was no longer present.

27

Main effect of Diet: (F1,74= 8.3 p=0.005) Main effect of Exposure: (F1,74= 7.2 p=0.009) 50 A C57BL6/J P70 B BALB/c P70 A B C 600 600

40)

* ) Std-H2O

m

m

(

(

DVD-H O

d 2

d

e

e

l

l l

30 400 l 400 Std-PEE e

The mice were placed along the edge of the walle and the number of head-dips in a 10

v

v

a a

r DVD-PEE

r

t

t

e

e c

20 200 min period was analysed. Each mouse was tested;c 200 one at a time, with all mice of a single

n

n

a

a

t

t

s

s

i

i D 10 strain being tested on the same day. ActivityD was recorded using video recording 0 0 software (Miglia TV) and the number of head-dips was manually recorded. The 0 Figure 2.3 Locomotionapparatus duringwas cleaned early between adulthood each mouse (P70). with 80 % ethanol. (A) Locomotion in C57BL/6J mice during adulthood (P70). Exposure to DVD deficiency as well as 15 C D PEE resulted* in hypoactivity. (B) Picture of open field arena (activity monitor). (C) Locomotion in BALB/c mice during adulthood (P70). There were no significant effects of Diet or Exposure.

(C57BL/6J Std-H2O n=19; DVD-H2O=24; Std-PEE n=8; DVD-PEE n=24. BALB/c Std-H2O n=36; 10 DVD-H2O n=30; Std-PEE n=20; DVD-PEE n=44). Data presented as mean  SEM.

2.3.4 Primer validation 5

qRT-PCR results and DNA gel electrophoresis confirmed the validity of the selected 0 primers and housekeeping gene as no dimers were observed and the molecular weight

was accurate with the expected weight (obtained from the original primer design- Cheng et al (2011)) (Fig. Fi2.g4)ure 6. The Holeboard. A. The holeboard apparatus with a C57Bl/6 mouse head-dipping into a hole in the floor. B. A schematic representation showing dimensions. The holeboard test was used to measure exploration based on the frequency of head-dipping.

2.5.4 Light/Dark Test 8 The light/dark test was used in the behavioural test battery to aid in the analysis of

anxiety levels of the mice (Crawley, 1985). The two-chambered light/dark apparatus 9 10 11 12 13 1 2 (3Fig 7)4 consisted5 6 of an7 open black box (30 cm high x 3014 cm x15 30 cm), with half of the

box containing a black insert (15 cm high x 30 cm x 15 cm) creating the dark chamber.

The insert had a small rounded open doorway in the centre leading to the light chamber.

The mice were placed in the doorway with their head inside the dark chamber and two

parameters were analysed. The time for the mouse to emerge from the dark chamber into

the light and the percentage of time the mouse spent in the light chamber in a 10 min period. Each mouse was tested individually, with all mice of a single strain being tested

on the same day. The test was recorded using a USB webcam and recording software

! "# #

28

Lane Sample Gene Lane Sample Gene 1 cDNA6 (1:5) Hprt1 9 cDNA7 (1:20) Vglut1 2 cDNA7 (1:5) Hprt1 10 cDNA6 (1:5) Vglut2 3 cDNA6 (1:20) Hprt1 11 cDNA7 (1:5) Vglut2 4 cDNA7 (1:20) Hprt1 12 cDNA6 (1:20) Vglut2 5 cDNA6 (1:5) Vglut1 13 cDNA7 (1:20) Vglut2 6 cDNA7 (1:5) Vglut1 14 cDNA6 (1:5) Vglut3 7 cDNA6 (1:20) Vglut1 15 cDNA6 (1:20) Vglut3 8 LADDER

Figure 2.4. Primer validation via DNA gel electrophoresis. Primers for Vglut1-3 were tested by qRT-PCR and visualized via DNA gel electrophoresis. No primer dimers were found and band were sitting at the correct molecular weight (Hprt1:165bp; Vglut1: 167bp; Vglut2: 173bp; Vglut3: 128bp) (Cheng et al 2011).

2.3.5 Vglut1-3 expression in early adulthood (P80-90)

The expression of Vglut1 and 3 was not significantly different as a result of treatment. There was a significant effect of strain, whereby BALB/c mice had higher levels of Vglut 3 compared to C57BL/6J (F1,92= 4.744 p<0.05; Figure 2.5 C, D). With regards to Vglut2, there was a significant three- way interaction Strain x Diet x Exposure (F1,89= 4.364 p<0.05; Fig. 2.6). C57BL/6J mice belonging to the Std-PEE group showed a significant downregulation in Vglut2 expression (t= 2.098, df: 15.389, p<0.05; Fig. 2.6A), while both DVD-H2O and DVD-PEE showed no difference in Vglut2 levels. On the other hand, BALB/c mice from the

Std-PEE group as well as the DVD-H2O group showed an upregulation of Vglut2 while the DVD-PEE animals showed no difference (Fig. 2.6B).

29

C57Bl/6J Vglut1 BALB/c Vglut1 A B Std-H2O 2.0 2.0

DVD-H2O

n

n

o

o i

i Std-PEE s

1.5 s 1.5

s

s e

e DVD-PEE

r

r

p

p x

1.0 x 1.0

e

e

e

e

v

v

i

i

t

t

a

a l

0.5 l 0.5

e

e

R R

0.0 0.0

C57Bl/6J Vglut3 BALB/c Vglut3 C D

2.0 2.0

n

n

o

o

i

i s

s 1.5 1.5

s

s

e

e

r

r

p

p x

x 1.0 1.0

e

e

e

e

v

v

i

i

t

t

a

a l

l 0.5 0.5

e

e

R R

0.0 0.0

Figure 2.5. Expression of Vglut1 and 3 in C57BL/6J and BALB/c hippocampal tissue. (A, B) There was no effect of treatment in the expression of Vglut1 in C57BL/6 or BALB/c adult hippocampal tissue. (C, D) Vglut3 expression was not affected by treatment in C57BL/6 or BALB/c hippocampal tissue, however, there was a main effect of strain, where BALB/c had a higher expression than C57BL/6J. (C57BL/6J Std-H2O n=12; DVD-H2O=12; Std-PEE n=7; DVD-PEE n=12. BALB/c Std-H2O n=12; DVD-H2O n=12; Std-PEE n=12; DVD-PEE n=12). Data presented as mean  SEM.

30

Main effect of Strain x Diet x Exposure: (F1,88= 4.364 p<0.005)

C57Bl/6J Vglut2 BALB/c Vglut2 A B Std-H2O 4 4 DVD-H O

* 2

n n

o Std-PEE

o

i i

s 3

s 3 s

s DVD-PEE

e

e r

r *

p

p x

2 x 2

e

e

e

e

v

v

i

i

t

t

a

a l

1 l 1 e

* e

R R

0 0 Figure 2.6. Expression of Vglut2 in C57BL/6J and BALB/c hippocampal tissue. (A) Vglut2 expression in C57BL/6 hippocampal tissue from the Std-PEE group was significantly reduced, while no difference was found in the DVD-H2O and DVD-PEE groups. (B) Vglut2 expression in BALB/c hippocampal tissue from the Std-PEE and the DVD-H2O groups was increased, while no difference was found in the DVD-PEE group. (C57BL/6J Std-H2O n=12; DVD-

H2O=12; Std-PEE n=7; DVD-PEE n=12. BALB/c Std-H2O n=12; DVD-H2O n=12; Std-PEE n=12; DVD-PEE n=12). Data presented as mean  SEM.

2.4 Discussion

Studies on DVD deficiency have shown behavioural and neurochemical alterations in adult offspring (Harms et al 2012a). Similarly, PEE models have reported alterations in both behaviour and gene expression (Sanchez Vega et al 2013, Zhang et al 2015). However, limited studies have explored the combination of both of these prenatal insults. The main findings from this study were: 1) C57BL/6J and BALB/c animals from the DVD-H2O and

Std-PEE showed increased locomotion compared to controls (Std-H2O); 2) mice from the ‘two-hit’ group (DVD-PEE) showed further hyperlocomotion compared to those under a singular exposure, suggesting a synergistic effect of DVD deficiency and PEE in locomotion at P21; 3) there was a main effect of Strain (C57BL/6J vs. BALB/c) on novelty- induced locomotion at P70. In C57BL/6J mice, there was a significant main effect of Diet (deplete vs. standard) and of Exposure (ethanol vs. water) on this measure, however, the pattern was different from results obtained at P21, as locomotion was decreased in prenatally exposed animals; 4) there was a significant effect of Strain x Diet x Exposure in the expression of Vglut2 in adult hippocampus.

31

2.4.1 Effects of DVD deficiency and PEE on early development

Gross developmental abnormalities were not expected in this experiment. Previous studies using this model of DVD deficiency have shown that Ca2+ levels are similar between control and DVD deficient offspring, and DVD-deficient mice attained all developmental milestones at the same stage as control mice (Harms et al 2012a). Similarly, given the low dose and brief period of exposure to ethanol in the model used in this experiment, typical developmental progression at different stages (P7, 14 and 21) was expected (Sanchez Vega et al 2013). Handling-induced cannibalism was a limitation in this experiment, as monitoring would ideally be performed at earlier ages (P7 and 14), however, this was limited to P21 to maximise litter numbers, necessary for further experiments.

Locomotion is a behavioural measure that is known to be affected as a result of both of these exposures individually and this finding was validated in the current experiment. Furthermore, the combination of both exposures (DVD-PEE) led to increased hyperlocomotion at P21, which is a novel finding. Vitamin D is known to have neuroprotective qualities (Alexianu et al 1998, Ibi et al 2001), therefore, its absence may exacerbate the effects of other exposures (such as PEE) targeting convergent pathways. For example, some have investigated vitamin D’s neuroprotective properties as a means to attenuate deficits caused by exposure to alcohol (Ngo 2013) and have reported that vitamin D is capable of ameliorating motor coordination, learning and memory deficits associated with developmental alcohol exposure (Idrus NM 2012), suggesting that the absence of vitamin D during critical developmental periods may lead to enhanced effects of secondary exposures (i.e. PEE). Another study looking at the combination of hypovitaminosis D and the effects of high fat diets on obesity and dopamine-related behaviours (Trinko 2013) found a significant increase in weight gain in animals that were vitamin D deficient and exposed to a high fat diet (HF-D) compared to those that received a full dose of vitamin D 6h prior to exposure to high fat diet (HF+D). Additionally, HF-D animals showed attenuated locomotor responses to amphetamine and increased oral amphetamine intake in contrast to HF+D animals. This provides evidence on the importance of the role vitamin D plays in the penetrance of second hit exposures.

32

2.4.2 Effects of DVD deficiency and PEE on adult behaviour

A significant effect of strain was encountered in novelty-induced locomotor behaviour, whereby C57BL/6J were significantly more active than BALB/c regardless of their experimental group, which is consistent with previous findings from previous studies examining differences between strains (Crawley et al 1997). Locomotion in C57BL/6J and BALB/c mice tested in a novel environment (activity monitor) at P70 was not consistent with the pattern of behaviour observed at P21. There was a main effect of Diet and of Exposure but the synergistic effect encountered at P21 was not found. C57BL/6J mice from the Std-PEE and DVD-PEE groups had a decrease in locomotion, which is inconsistent with results from previous studies using the same PEE mouse model (Sanchez Vega et al 2013, Zhang et al 2015) where adult hyperlocomotion was the main finding. However, contradictory results at different ages and from different studies is not uncommon. For example, a longitudinal study in rats encountered hyperlocomotion in PEE adolescent rats and a shift in direction (hypolocomotion) during adulthood (Bond et al 1977). Some of the factors that may lead to these differences are 1) repeated testing; mice tested for locomotion during prepubescence were also tested as adults. There are a number of studies that have shown that repeated handling and testing affects the behavioural phenotype of the animals (Gariépy et al 2002). To address this, separate cohorts may need to be tested at P21 and at P70. 2) weaning age and housing environment: animals from the Zhang (Zhang et al 2015) study were weaned at P21 and individually housed in open top cages until adulthood, while those in the current study were weaned a week later (P28), and group housed in filtered cages. There is evidence to suggest that isolation, different housing environments and noise levels have an impact on behaviour and may interfere with the replication of previously observed phenotypes (Logge et al 2014). Furthermore, the findings reported in this experiment show that DVD deficiency and PEE have a short and long-term effect in locomotor behaviour, and the contrasting direction of change may be due to one or a combination of the environmental parameters discussed above.

2.4.3 Prenatal exposures and gene expression

The expression of Vglut1-3 was evaluated in adult hippocampal samples from each experimental group. While there were no significant effects of Diet or Exposure on expression levels of Vglut1 or 3, there was a significant interaction of Strain x Diet x

33

Exposure on the expression of Vglut2. Std-PEE C57BL/6J mice showed a downregulation in Vglut2, while Std-PEE and DVD-H2O BALB/c had an increase in Vglut2 expression. Previous PEE studies have shown alterations in the epigenetic regulation of Vglut2, which affects its adult expression in C57BL/6J mice (Zhang et al 2015). The present findings in BALB/c mice are novel. Furthermore, the direction of change in this study are opposite to those previously reported, although it is consistent with the behavioural findings discussed above. The internal uniformity of these results suggests that housing conditions, for example, may have led to the directional change. Given that Vglut2 is developmentally regulated, and handling at early stages (P7, P14, P21) can be stressful for pups, stress hormones may have interfered with the effects of PEE on the regulation of Vglut2, resulting in contradictory results as observed in adulthood. One way to address in future studies would be to test the expression of Vglut2 at early developmental stages in PEE animals and test a separate cohort during adulthood. Additionally, further work on the effects of stress hormones (corticosteroids, altered hypothalamic-pituitary-adrenal (HPA) axis) on the epigenetic regulation of Vglut2 would be valuable to better understand their relationship.

Expression of Vglut1 and 3 showed greater variability in both strains, while Vglut2 was less variable. This may be due to a number of factors, including the expression levels of each of these genes in relation to tissue type and age. For example, Vglut3 expression was much lower than Vglut2 irrespective of experimental group, this was noted during experimental procedures where the average Ct value (reflective of number of PCR cycles required for the fluorescent signal to cross the threshold, which denotes a sample as ‘positive’ or above background) for Vglut3 was high, which may compromise the test’s sensitivity and would reduce accuracy. Overall, these findings reiterate previous reports showing stable expression of Vglut2 in adult hippocampal tissue compared to Vglut1 and 3. 2.4.4 Conclusion

The results obtained from this experiment were an initial step into understanding the effects of the combination of DVD deficiency and PEE on brain function and behaviour with two main outcomes. Firstly, given that BALB/c mice did not show a clear vulnerability compared to C57BL/6J in the recorded measures, the latter was selected to proceed with future experiments, considering its well-known genetic background and the availability of supporting literature on environmental exposures compared to BALB/c. Secondly, adult

34 behaviour and hippocampal gene expression results were subtle, but suggested hippocampal-dependent memory and glutamatergic signalling may be affected as a result of DVD deficiency and PEE. Therefore, the aim of the following experiments was to assess learning and memory, and probe glutamatergic regulation in adult offspring.

35

Behavioural phenotype of adult C57BL/6J mice exposed to DVD 3 deficiency and PEE

36

Chapter 3. Behavioural phenotype of adult C57BL/6J mice exposed to DVD deficiency and PEE.

3.1 Introduction

Vitamin D has been associated with altered cognition in adults. Clinical studies have reported significant positive associations between low serum 25(OH)D3 concentrations and low scores on tests that evaluate global cognitive function such as Mini Mental State Examination (MMSE), Clinical Dementia Rating (CDR) and Short Blessed Test (SBT) (Przybelski et al 2008, Wilkins et al 2006). However, findings have not been consistent across studies, for example, a systematic review found three studies showing positive associations between serum 25(OH)D3 concentrations and global cognitive functions, and three other studies where 11 non-significant associations with specific aspects of cognition were found. (Annweiler et al 2009). The inconsistencies found in these studies are likely due to methodology heterogeneity and lack of control of confounders. For this reason, animal models have been a useful tool to investigate the association of vitamin D deficiency with adult behaviour, as experimental conditions can be controlled, thereby reducing the number of confounding factors.

DVD deficiency in mice has been shown to affect long-term behaviour in a strain- dependent manner. Spontaneous hyperlocomotion was found in 129/Svj mice, but not in C57BL/6J mice, and increased exploration in both strains, as measured by the frequency of head dipping on the hole board test (Harms et al 2008a). With regards to cognition, impairments in hippocampal-dependent memory in adult DVD-deficient C57BL/6J mice have been reported in a learning test based on olfactory cues (Fernandes de Abreu et al 2010). Subtle alterations in response inhibition were found in C57BL/6J DVD-deficient mice during the 5-choice continuous performance test, which suggests DVD deficiency may disrupt systems specific for compulsive or reward-seeking behaviours (Harms et al 2012b).

The effects of DVD deficiency on the regulation of glutamate and dopamine systems has been previously assessed via pharmacological investigations, using psychomimetic drugs such as MK-801 and amphetamine, where hyperlocomotor response is used as a behavioural readout. MK-801 response was not affected by DVD deficiency in C57BL/6J

37 or 129X1/Svj mice, while sensitivity to amphetamine was altered in 129X1/Svj mice, where males showed enhanced sensitivity and females a reduced locomotor response (Harms et al 2012a). By contrast, DVD deficiency resulted in increased MK-801-induced hyperlocomotion in male rats (Kesby et al 2006), and enhanced amphetamine sensitivity in female rats (Kesby et al 2010), which suggests that this response is dependent on background strain, species and sex.

Similar to findings from DVD deficiency studies, the effects of PEE on adult behaviour are also influenced by a number of factors including species, background strain, dosage and timing of exposure. It is widely accepted that exposure to high doses of ethanol has long- lasting detrimental effects on brain development and behaviour (locomotion (Becker et al 1989), sociability and learning and memory (Marquardt et al 2016)), whereas the effects of moderate doses are not fully understood, even though structural and behavioural defects have been shown in humans (Sood et al 2001) and animal models (for example Brolese et al (2014), Hamilton et al (2014)). Furthermore, the hippocampus is one of the areas of the brain that are most vulnerable to the effects of ethanol (Guerri 2002), and moderate PEE has been shown to enhance the performance of PEE male mice in the MWM (Sanchez Vega et al 2013) or have no effect in rats (Cullen et al 2013).

High doses of ethanol exposure have been shown to induce alterations in glutamatergic signalling, along with disruptions in hippocampal synaptic plasticity, including dysregulation of Vglut1, Vglut2, complexin 1, various NMDA receptor subunits and EAAT1 and 3 (Kaminen-Ahola et al 2010b, Naassila et al 2002, Zhang et al 2015, Zink et al 2009, Zink et al 2011). Moreover, a study in zebra fish showed that ethanol exposure at various doses impaired cerebral glutamate transport in adult fish, contributing to long-term alterations in the homeostasis of glutamatergic signalling (Baggio et al 2017). Another study in rats showed that PEE treatment significantly decreased the number of [3H]MK-801 binding sites (NR2A subunits) (Vallés et al 1995), which may be associated with the cognitive deficits observed in PEE rats.

Considering the subtle behavioural effects resulting from DVD deficiency and moderate PEE, the aim of this chapter was to study whether the combination of both exposures would result in a more prominent behavioural phenotype, focusing on locomotion and hippocampal-dependent learning and memory, and by a psychomimetic challenge using

38 the non-competitive NMDA antagonist MK-801 to probe the regulation of glutamatergic signalling.

3.2 Materials and methods

3.2.1 Animals, housing and exposures

Forty adult male and 60 four-week old female C57BL/6J mice were purchased (ARC, Perth, WA) and housed as described in section 2.2.1. The first 20 sires were used throughout the first two waves to reduce variability from different sires and the second group of 20 was used with the last cohort of females. Dams were randomly allocated to one of four experimental groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) following diet and exposure methods described in section 2.2.2 and 2.2.3.

3.2.2 Adult locomotion

Each mouse was placed in a clear open field (27.5 x 27.5 x 30cm, Med Associates Inc, USA) within a sound attenuated chamber and activity levels were recorded for 30 min. The light level was set at 18 lux. As a measure of spontaneous activity, distance travelled was calculated using activity monitor tracking software based on beam breaks from three 16 beam infrared arrays and was sampled in 1 min time bins (Walsh et al 1976).

A

Figure 3.1. Picture of open field arena (activity monitor).

39

3.2.3 Active place avoidance (APA)

The active place avoidance (APA) task is a hippocampus-dependent spatial learning task (Cimadevilla et al 2001, Wesierska et al 2005). The apparatus (Bio-Signal Group) consisted of an elevated arena with a grid floor fenced with a 32-cm-high transparent circular boundary (enclosed arena diameter 77 cm), located in a room with visual cues on the walls. The arena rotated counter clockwise (1 rpm) and an electric shock could be delivered through the grid floor, which also rotated. The position of the animal in the arena was tracked using an overhead camera linked to Tracker software (Bio-Signal Group). During trials, a mouse was placed in the arena and trained to avoid a 60° shock zone, the positioning of which was kept constant (i.e., did not rotate) in relation to the room coordinates; the mouse's “start” position was always near the wall on the side of the arena opposite the shock zone. Entrance into the shock zone led to the delivery of a brief foot shock (500 ms, 60 Hz, 0.5 mA). If, after the initial shock, the animal remained in the shock zone, further shocks were delivered at 1.5 s intervals until the animal moved out of the zone. Each training session lasted 10 min (habituation sessions were 5 min long), and recorded tracks were analysed offline using Track Analysis software (Bio-Signal Group). Mice were habituated to the training environment 24 h before the initial experiment (i.e., a mouse was placed in the rotating arena with the shock turned off and allowed to explore the arena for 5 min).

Figure 3.2. Active Place Avoidance. Picture of the APA testing arena. (B) Spatial cues used on each wall surrounding the testing arena.

40

3.2.4 MK-801 locomotor response

The drug used to assess psychomimetic-induced hyperlocomotion was MK-801 (Sigma, MO, USA). It was diluted to a concentration of 0.1 mg/ml in 0/9% SAL. Stock solutions were stored at -20C. Mice were given intraperitoneal injections at a volume of 5 ml/kg, resulting in a dose of 0.5 mg/kg MK-801. This dose was based on other studies and typically elicits a high locomotor response in mice, with little impact from stereotypic or ataxic behaviours (van den Buuse 2009, Wu et al 2005). 0.9% SAL was used as vehicle, injected at a volume of 5 ml/kg. Each mouse had its own vehicle/SAL control, so that the baseline level of locomotion for each animal was used to control for drug-induced locomotion.

On the day of testing, mice were transported into the behavioural testing room, weighed and habituated to the testing room for at least an hour before testing. Mice were habituated to the activity monitor testing arena for 30 min. Mice were then injected I.P. with SAL and placed back in the activity monitor for a further 30 min. Lastly, mice were injected I.P. with 0.5 mg/kg MK-801 and placed in the activity monitor for 120 min.

3.2.5 Statistical Analysis

Results were analysed for statistical significance using the SPSS statistics software package (ver. 24, SPSS Inc., Chicago, Illinois). Where no significant Group x Sex interactions were encountered, data were presented pooled for sex. Data were analysed using ANOVA and, where appropriate, repeated measures were used. Student’s t-test was used to assess the difference between two groups. Associations between behavioural results and gene expression levels were performed using non-linear regression. A p value of 0.05 or less was considered to be statistically significant.

41

3.3 Results

3.3.1 Adult locomotion

Novelty-induced locomotion was evaluated from a total of 112 adult mice (61 from Wave 4 and 51 from Wave 5) representing all groups (DVD-PEE: 15 males, 12 females; DVD-

H2O: 18 males, 21 females; Std-PEE: 16 males, 10 females; Std- H2O: 8 males, 12 females) in an activity monitor for 30min. There was a significant main effect of Sex, whereby males were more active than females (F1,108= 6.8, p=0.011; Fig. 3.3). There was also a significant main effect of Exposure, whereby PEE animals travelled less distance compared to those from the water groups (F1,108= 11.7, p=0.001; Fig. 3.3) across the

30min trial, and a significant interaction of Diet x Exposure (F1,108= 4.01, p=0.048). The pattern of behaviour was similar in males and females; however, independent sample t- test (Dunnett’s test) comparing Std-H2O to each exposure group, split by sex, revealed that only male PEE mice were significantly hypoactive compared to controls (t=3.510, df=8.904, p=0.007, Fig. 3.3 A), while the activity levels in PEE females did not reach significance (t=2.01, df=20, p=0.052, Fig. 3.3 B).

Males Females A B Std-H2O 10000 10000

DVD-H2O

n

n i

i 8000 8000 m

m Std-PEE

0

0

3

3

)

) 6000 DVD-PEE 6000

m

m c

c *

(

(

e

e 4000 4000

c

c

n

n

a

a

t

t

s

s i

i 2000 2000

D D

0 0

Figure 3.3. Locomotion during early adulthood (P70).

Locomotion in C57Bl/6J mice during adulthood (P70). (A) Std-PEE males were significantly hypoactive compared to controls (Std-H2O). Males from the DVD H2O or the DVD-PEE did not show significantly altered locomotion. (B) There was no significant effect of Diet or Exposure in females. Data are presented as mean ± SEM. * p< 0.05 between control and Std-PEE male mice.

42

3.3.2 Active place avoidance

All animals learned the task across a 4-day protocol at P71-75. Animals from all groups learned to avoid the shock zone using spatial cues placed in the room, which was shown by a progressive decrease in total shocks received and therefore an increase in time spent outside the shock area, therefore, there was a main effect of Day on the number of shocks received (F1,108=58.2, p=0.000). No significant effect of Diet, Exposure or Sex was detected in any of the parameters assessed in this task, including latency to first shock, total number of shocks per trial, total path, speed, maximum time avoiding the shock zone and time to second entry into shock zone (Males Fig. 3.4A-F; Females Fig. 3.5A-F).

A subset of males from all experimental Groups (n=8/Group) were tested at older age (P210) in the same task, using different visual cues. The task was acquired by all animals, shown as a significant effect of Day in the time spent avoiding the shock zone

(F1,28=213.9, p=0.000) and in the number of shocks received (F1,108=94.1, p=0.000). There was no significant effect of Diet or Exposure in any of the parameters measured in this task, as described above (Fig. 3.6A-F)

43

P70 Males A B C 45 15

300 Std-H2O DVD-H2O

40 Std-PEE

k

c )

s DVD-PEE

o

m k

h 10

c

c

s

(

200 o

t

h

h

s

t

r

S

i a

35

f

f

P

o

o

l

t

.

a

t

o

e

o

N

m T i 5 T 100 30

0 0 25 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

D E F 7.0 400

300

6.5

) s

300 (

)

y

s

r

(

6.0 t

)

d

n

i

s

/

E

o

v m

d 200

c

A

n

(

o

5.5 e 200

d

c

e

m

e

i

e

S

T

p

.

o

S

x

t

5.0 a e

M 100 m

100 i T 4.5

4.0 0 0 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

44

Figure 3.4. Spatial memory in the APA test in adult male mice. Male mice from all groups progressively learned to avoid the shock zone, demonstrated by the increase in time to first shock and by the reduction in total number of shocks received in males (A, B). Other parameters measured included total path (C), speed (D), . time avoiding shock zone (E) and time to second entry (F). No significant effect of Diet or Exposure was found across the four-day task. Data presented as mean  SEM.

45

P70 Females A B C 45 15

300 Std-H2O DVD-H2O

40 Std-PEE

k c

) DVD-PEE

s

o

m k

h 10

c

c

s

(

200 o

t

h

h

s

t

r

S i

a 35

f

f

P

o

o

l

t

.

a

t

o

e

o

N

m T i 5 T 100 30

0 0 25 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

D E F 7.0 400 400

6.5

) s

300 ( 300

)

y

s

r

( t

6.0

)

d n

i

s

/

E

o

v

m

d

c

A

n

(

o

5.5 e 200 200

d

c

e

m

e

i

e

S

T

p

.

o

S

x t

5.0 a

e

M m

100 i 100 T 4.5

4.0 0 0 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

46

Figure 3.5. Spatial memory in the APA test in adult female mice. Female mice from all groups progressively learned to avoid the shock zone, demonstrated by the increase in time to first shock and by the reduction in total number of shocks received in males (A, B). Other parameters measured included total path (C), speed (D), max. time avoiding shock zone (E) and time to second entry (F). No significant effect of Diet or Exposure was found across the four-day task. Data presented as mean  SEM.

47

A B P210 Males C 400 15 40

Std-H2O

DVD-H2O

300 35 Std-PEE

k c

10 ) DVD-PEE

s

o

m

k

h

c

c

s

(

o

t

h

h

s

t

r

S

i a

200 30

f

f

P

o

o

l

t

.

a

t

o

e

o

N

m T

i 5 T 100 25

0 0 20 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

D E F 6.5 500 500

6.0

400 400

)

s

(

)

s

y

(

r

5.5 t

)

d

n

i

s

/ E o 300

300

v

m

d

c

A

n

(

e

5.0 o

d

c

e

m

e

i

e

T

S

p

200 200

.

o

S

x

t

4.5 a

e

M

m i 100 T 100 4.0

3.5 0 0 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4 Day 1 Day 2 Day 3 Day 4

48

Figure 3.6. Spatial memory in the APA test in older adult male mice. Males from all groups were re-trained at older age, all mice progressively learned to avoid the shock zone, demonstrated by the increase in time to first shock and by the reduction in total number of shocks received (A, B). No significant effect of Diet or Exposure was found across the four-day task on any of the parameters assessed (A-F). Data presented as mean  SEM.

49

3.3.3 Psychomimetic-induced locomotion

Adult male offspring were treated with MK-801 and locomotor response was assessed. Mice from all Groups habituated to the activity monitor (baseline activity), there was a slight increase in activity post-SAL injection, which was normalized (back to baseline) within 30min, and further hyperlocomotion was found upon MK-801 injection. Data was analysed in six 30min blocks, split into six 5min bins each; block 1 corresponds to the 30min habituation trial, block 2 to SAL trial and blocks 3-6 to MK-801 response. There was a significant effect of Block (F5,24=56.74, p<0.001) and Bin (F5,24=11.22, p<0.001), whereas Diet or Exposure had no significant effect on the locomotor response of C57BL/6J mice to

MK-801 (Diet effect F1,28= 0.134, p=0.71; Fig. 13; Exposure effect F1,28=0.547, p=0.46), and there were no significant interactions between Diet and Exposure (F1,28=1.551, p=0.22, Fig. 3.7). Hyperlocomotor response to MK-801 was also analysed at peak activity, which in this case was 60min post MK-801 administration. Results showed no significant main effects of Diet (F1,28= 0.005, p=0.944), Exposure (F1,28= 1.223, p=0.274) or

DietxExposure (F1,28.739, p=0.397) during this time point either.

Males )

n 2500 i Std-H2O

m Saline MK-801 5

/ 2000 DVD-H2O

m

c ( Std-PEE

d 1500

e l

e DVD-PEE v

a 1000

r

t

e c

n 500

a

t

s i

D 0 -60 -30 0 30 60 90 120 Time (min)

Figure 3.7. MK-801 hyperlocomotor response in adult male mice. There was no significant effect of Diet or Exposure in MK-801 induced hyperlocomotion in C57BL/6J males. Data presented as locomotor response including 30min habituation to activity monitor (-60 to -30min), 30min post-saline administration (-30 to 0min) and 120min post MK-801 IP injection (0-120min), for males of all Groups.

50

3.4 Discussion

The findings from this experimental chapter provide no evidence to suggest that DVD deficiency and PEE act upon convergent pathways that result in exacerbated alterations regarding adult behaviours relevant to hippocampal-dependent learning and memory and glutamatergic signalling. Furthermore, results from this study showed: 1) hypolocomotion in Std-PEE adult males, 2) unaffected learning and memory performance all Groups at P70 or in males at P210, and 3) unchanged response to MK-801 of male offspring as a result of Diet or Exposure.

Table 3.1. Summary of Behavioural Phenotype in Adult Offspring.

Test DVD- H2O Std-PEE DVD-PEE ♂ ♀ ♂ ♀ ♂ ♀

Locomotion - - - - -

Learning and ------memory Psychomimetic ------response

3.4.1 Effects of DVD deficiency and PEE on adult locomotion

Consistent with (Harms et al 2008a), the present study showed that DVD deficiency did not affect adult locomotion in response to novelty, while PEE resulted in hypoactivity, which is in agreement with findings from the previous chapter (Chapter 2) using a separate cohort and with a different study using the same model of exposure (Zhang 2013). The combination of DVD deficiency and PEE did not result in an exacerbated phenotype as hypothesised. On the contrary, the activity levels were comparable to controls in the DVD- PEE group, suggesting these exposures did not act upon convergent pathways. The hypoactivity found in PEE males may reflect alterations in glutamate regulation, given that ethanol exposure has been previously shown to dysregulate the expression of NMDA

51 receptor subunits (Zink et al 2011) as well as glutamate transporters in the hippocampus of male adult mice (Zhang et al 2015).

3.4.2 Effects of DVD deficiency and PEE on learning and memory

There were no alterations in learning and memory as measured by the active place avoidance in adult animals exposed to DVD deficiency or PEE. This is the first study to use the APA task to asses hippocampal-dependent learning and memory using these developmental exposures, and there are no previous data with which to compare these results. A study in mice found impaired learning in a hippocampal-dependent task based on olfactory cues at 30 weeks of age, where DVD-deficient mice made significantly less correct responses on the last day (5) of training compared to controls (Fernandes de Abreu et al 2010). Meanwhile, a study in DVD-deficient rats reported very subtle alterations in learning and memory, showing an impairment of latent inhibition, whereas spatial learning (Y maze) or two-way active avoidance learning remained unaffected (Becker et al 2005).

With regards to ethanol exposure, learning and memory was altered in humans and rats (Mattson et al 1999), particularly with high doses of ethanol. However, the effects of moderate levels of exposure are not as consistent; a previous study in rats found no alterations in the MWM (Cullen et al 2013), while another study in mice showed an enhanced performance in PEE males using the same task (Sanchez Vega et al 2013). Moreover, the results from the present study showed no effects of PEE, even though the same model of exposure was used. The discrepancy between these results may be due to the differing nature of the tasks, where the MWM induces high levels of stress and requires a significant amount of motor activity (swimming), while the APA requires locomotion but does not have the added stress of swimming (Lesburguères et al 2016); perhaps the credit found in ethanol-exposed animals in the MWM was driven by stress and not particularly due to superior learning skills, while the settings on the APA were not challenging enough to uncover an impairment. If the test was to be repeated, the shock strength could be lowered (to reduce stress-driven performance), the shock zone could be smaller and spatial cues shuffled on each day of testing. Studies comparing the MWM and APA have reported inconsistent findings across tasks, some arguing the APA is a more sensitive spatial learning task than the MWM (Lobellova et al 2013), while others suggest the opposite (Stuchlik et al 2004).

52

As previously mentioned, a study on DVD-deficient mice reported significant hippocampal loss in older mice, along with impaired learning and memory, which suggested the effects of DVD deficiency may be more prominent as animals age (Fernandes de Abreu et al 2010). In addition, a study focusing on the effects of early stress (ES) on hippocampal- dependent memory reported biphasic changes in performance in the MWM. Young adult ES rats displayed enhanced hippocampal-dependent memory compared to controls, while the opposite was found in middle aged ES rats (Suri et al 2013). Therefore, in the current study, males from both DVD and PEE groups were re-trained in the APA at P210, however, animals from all groups were able to learn the task, suggesting the impact of both exposures was not exacerbated with increasing age.

3.4.3 Effects of DVD deficiency and PEE on psychomimetic-induced locomotion

MK-801, a non-competitive NMDA receptor antagonist, was used to challenge glutamatergic signalling in adult males. There was a significant effect of Exposure in locomotion during the habituation period, which is consistent with results from our previous experiment, showing Std-PEE males were hypoactive compared to controls. Nevertheless, there was no significant effect of Diet or Exposure on the hyperlocomotor response observed after MK-801 administration, which suggests that changes observed in the baseline activity levels of PEE may not be due to aberrant glutamatergic regulation but rather a different neurotransmitter signalling pathway that can also influence locomotor response, such as GABA. Multiple studies have reported alterations in the GABA system of adult offspring exposed to single or multiple doses of ethanol during development, however, the underlying mechanisms for GABA dysregulation and altered behaviour are not known (Valenzuela et al 2011). A single ethanol dose during GD8 resulted in elevated levels of 5 GABAA in the brains of adult mice (Toso et al 2006, Webster et al 1983). Additionally, repeated exposure to high ethanol concentrations throughout pregnancy has been shown to induce persistent alterations in GABA transmission and increased levels of

GABAA receptor subunits in the cortex and hippocampus of adult guinea pigs (Bailey et al 2001). This evidence suggests GABAergic signalling is another potential neurotransmitter system of interest in relation to PEE and its long-term behavioural effects, and this should be investigated in future studies.

53

3.4.4 Conclusion

The main findings from this chapter do not support the hypothesis that DVD deficiency exacerbates the effects of PEE on adult behaviour. In summary, the results obtained in this experiment showed that PEE alone has an effect on male adult locomotion, while DVD deficiency, PEE or their combination resulted in no significant dysregulation of adult hippocampal-dependent learning and memory or psychomimetic-induced locomotion. The locomotor finding on PEE males was consistent with previous literature, where PEE resulted in altered activity levels (Kleiber et al 2011, Zhang 2013). However, a number of findings were not consistent with previous literature, or as hypothesized, including the effects of DVD and PEE on learning and memory and the locomotor response to MK-801. This may be due to differences in experimental settings compared to previous studies, such as housing conditions and repetitive handling, or due to technical inconsistencies, such as the efficacy of dietary manipulation, or ethanol consumption.

54

Interaction of DVD deficiency and PEE on maternal metabolism and neonatal gene 4 and protein expression

55

Chapter 4. Interaction of DVD deficiency and PEE on maternal metabolism and neonatal gene and protein expression.

4.1 Foreword

This chapter consists of two studies. The first study considers metabolism of ethanol in vitamin D deplete and control dams to study whether maternal vitamin D status interferes with the dams’ efficiency to metabolise ethanol, which would affect the level of exposure in the offspring. The second study examined brain tissue from offspring on the day of birth for broad screening of gene and protein expression. This is a crucial time point to assess the effects of DVD deficiency and PEE because it occurs prior to vitamin D reintroduction and excludes potential postnatal confounding factors, such as maternal care and housing conditions.

4.2 Influence of maternal vitamin D status on alcohol metabolism

4.2.1 Introduction

Multiple studies in humans have reported that one of the side effects from chronic alcohol exposure is vitamin D deficiency, as the toxic effects of alcohol impairs vitamin D/calcium homeostasis and results in decreased bone mineral density (Laitinen et al 1991, Mercer et al 2012, Sampson 1997). Moreover, alcohol metabolism is taxing on the liver, which also metabolises vitamin D to its active form. However, to the best of my knowledge there are no studies looking at the reverse situation, metabolism of alcohol in vitamin D deficient patients.

Vitamin D is synthesized from 7-dehydrocholesterol within the skin via UVB radiation. The amount of synthesis is dependent on a number of factors including season, latitude, skin pigmentation, age, time spent outdoors, sunscreen use and type of attire worn during exposure to sunlight (Chen et al 2007, Holick 1995). The conversion of 7- dehydrocholesterol in the skin of humans and animals, forms vitamin D3 (Lehmann et al 2010). Vitamin D is converted to its biologically active form via two enzymatic steps, the first of which occurs in the liver. Vitamin D is hydroxylated to 25(OH)D3 via either the

56 microsomal (CYP2R1) or the mitochondrial (CYP24A1) P450 25-hydroxylase enzymes

(Schuster 2011). 25(OH)D3 is then converted to the biologically active, 1,25- dihydroxyvitamin D (1,25(OH)2D3) in the kidney via 1α-hydroxylase (CYP27B1). 1α- hydroxylase is tightly controlled via feedback mechanisms from parathyroid hormone, calcium, phosphate, calcitonin, fibroblast growth factor 23, and vitamin D itself (Lehmann et al 2010).

Alcohol dehydrogenase (ADH) and cytochrome P450 are the major enzyme systems responsible for the oxidation of ethanol, and they are both present to the largest extent in the liver (Khanna et al 1980). Even though ethanol is a nutrient and has a caloric value, it cannot be stored and utilized in the same way as carbohydrates and lipids, therefore it remains in body water until eliminated. Additionally, unlike major nutrient metabolism, alcohol is under little to no hormonal control, therefore, its oxidation takes preference over other nutrients, putting the liver under a major burden to oxidize alcohol for elimination from the body (Zakhari 2006). Furthermore, the rate of alcohol metabolism is faster in animals with small body weight compared with larger animals. For example, mice metabolise alcohol 5 times faster than humans. These alcohol metabolism rates are correlated with the basal metabolic rate for each species (Addolorato et al 1997, Lands 1995, Lieber 1991, Morgan et al 1988, Salaspuro et al 1978). Alcohol elimination rates in very young animals are low because ADH and CYP2E1 are not fully expressed, which means that the foetal liver does not have an efficient alcohol elimination rate and may have detrimental consequences (such as FASD).

Therefore, the main aim of this study was to analyse whether maternal ethanol metabolism was altered in vitamin D deficient dams, whereby the absence of vitamin D during ethanol exposure interfered with the ethanol exposure, which may explain the diminished effect seen in the DVD-PEE group compared to Std-PEE in the previous chapter.

57

4.2.2 Materials and methods

4.2.2.1 Animals, housing and exposures

Twenty adult male and 40 four-week old female C57BL/6J mice were purchased (ARC, Perth, WA) and housed as described in section 2.2.1. The same sires were used throughout the two cohorts of females to reduce variability from different sires. Dams were randomly allocated to one of four experimental groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) following diet and exposure methods described in section 2.2.2 and 2.2.3.

4.2.2.2 Maternal blood sampling and tissue collection

Body weight measurements were recorded and terminal blood samples were collected from two waves of dams (20 dams each) using a 25-gauge needle and a 1ml syringe via cardiac puncture in the morning of the last ethanol-consumption day (GD8). Blood samples were centrifuged at 5000rpm for 10min to obtain sera, which was then kept frozen at -80C until time of experiment. Liver tissues were retrieved immediately after blood collection and snap frozen until further analyses. The second wave of dams (20 females) underwent an oral gavage of 10% ethanol solution 30min prior to terminal bleed to provide a positive control for ethanol exposure versus voluntary drinking volumes.

4.2.2.3 Maternal serum 25(OH)D levels

Sera collected from all dams from Waves 1 and 2 (n=20/Diet) was used to measure the levels of 25(OH)D via liquid chromatography-tandem mass spectrometry on a 4000 QTrap API AB mass spectrometer using a previously described procedure for assaying dried neonatal blood spots (Kvaskoff et al 2016, Kvaskoff et al 2012).

4.2.2.4 Maternal blood alcohol concentration (BAC) determination

Maternal blood ethanol content resulting from self-administration of a 10% ethanol in water solution or water only was determined enzymatically. Serum samples were mixed with an alcohol reagent (A7504-39; Pointe Scientific) and assayed immediately using a spectrophotometer (CLARIOstar plate reader, BMG Labtech) at 340nmwavelength. Blood ethanol standards were created by mixing alcohol standard (104 mg/dl) with alcohol

58 reagent and immediately assayed. Each sample was analysed in duplicate and experimental samples were compared against a standard curve established by mixing alcohol standard with various ethanol concentrations (0%, 0.025%, 0.05%, 0.1%) to determine blood ethanol concentrations. Control (Std-H2O) dams were used as negative controls. Differences in BAC between ethanol-treated and control dams were analysed using a t-test. Statistical significance was set at p < 0.05. Ethanol concentration was calculated using the following formula:

Abs sample Ethanol concentration (mg/dL)= ( ×[Standard]) ×-1 Abs Standard

4.2.2.5 Alcohol dehydrogenase (ADH) levels in maternal liver tissue

Liver tissue samples from a total of 40 dams (15 DVD-PEE, 15 Std-PEE, 5 DVD- H2O, 5

Std- H2O) were prepared according to the manufacturer’s instructions (Sigma Aldrich MAK053-1KT) and assayed in a 96-well plate. Briefly, samples were homogenized in ADH assay buffer, then diluted to fit within the linear range of the standard curve (made up of multiple dilutions of a 1mM NADH standard solution). Reaction mixes were set up by combining ADH assay buffer, developer and 2M ethanol and adding that to 50ul of diluted sample. Plates were incubated at 37C and absorbance was read every 5min at 450 nm. ADH activity was calculated using the equation: ADH Activity= (B x Sample Dilution Factor)/(reaction time x V), where B was the amount (nmole) of NADH generated between the initial and final read, and V was the sample volume (ml) added to well. ADH activity was reported as nmole/min/ml= milliunit/ml

4.2.3 Results

4.2.3.1 Maternal body weight and serum 25(OH)D levels

Body weight assessment showed that there was no significant effect of Diet (F1,34= 1.01, p=0.32, Fig. 4.1 A) or Exposure (F1,34= 1.78, p=0.19, Fig. 4.1 A) on this measure. Serum vitamin D levels were measured in dams from two separate cohorts. There was a significant difference in 25(OH)D levels between Std and DVD groups, irrespective of

Exposure (H2O or PEE) (F1,34= 157.48, p<0.0001, Fig. 4.1 B), whereby dams on the Standard diet had significantly higher levels of vitamin D compared to those from the Deplete diet group.

59

Maternal body weight (8.5dpc) Maternal vitamin D levels A B 25 100

) * * M 80 20 n Std-H O

( 2

]

)

D g

( DVD-H O

15 H 60 2

t

O

h

5 g

i Std-PEE

2 [ e 40

10 W

m DVD-PEE

u r

5 e 20 S 0 0

Figure 4.1. Maternal body weight and serum 25(OH)D levels (A) There was no significant effect of Diet or Exposure in maternal body weight, recorded at 8.5dpc. (B) Levels of 25(OH)D were assayed using dam sera. A significant effect of Diet was found, showing dams on Std diet had significantly higher levels of vitamin D compared to those from the deplete group. Data presented as mean  SEM, *p<0.05 (n=10/Group).

4.2.3.2 Maternal blood alcohol concentration

The concentration of alcohol was measured in a total of 28 dams at GD8. Half of the dams received an acute dose of 10% ethanol (via oral gavage) 30min prior to blood collection to use as positive control, in case dams had not drank from their bottles shortly before blood sampling. There was a significant effect of Gavage, whereby dams who received acute alcohol administration had a significantly higher blood alcohol concentration (BAC) than those in the non-gavage group (F1,27= 41.939, p<0.0001, Fig. 4.2). There was no significant effect of Diet in BAC (F1,27=0.98, p=0.33).

60

Maternal Blood Alcohol

0.4 Std-PEE DVD-PEE

0.3

L

d

/

g u

0.2

C

A B 0.1

0.0 No Gavage Gavage

Figure 4.2. Blood alcohol concentration in dams at GD8. Dam serum was used to measure the concentration of alcohol on the last day of ethanol exposure. No significant effect of Diet was found on this measure. Dams that received an acute dose of 10% ethanol 30min prior to blood collection showed a significantly higher concentration of ethanol compared to those form the ad-libitum groups. (n>6/Group).

4.2.3.3 Maternal liver Alcohol Dehydrogenase levels

Liver tissue collected from 35 dams at GD8 was prepared for measuring total levels of

ADH. No significant effect of Diet (F1,34=1.89, p=0.18) or Gavage (F1,34=2.341, p=0.14) was found for this measure (Fig. 4.3).

Maternal Alcohol Dehydrogenase

20

Std-PEE )

l DVD-PEE m

/ 15

n

i

m

/

e l

o 10

m

n

(

H 5

D A

0 No Gavage Gavage

Figure 4.3. Liver ADH levels in dams at GD8. ADH levels were measured in maternal liver tissue collected on the last day of ethanol exposure (GD8). No significant effect of Diet or Gavage were found on this measure (n>8/Group).

61

4.2.4 Discussion

The aim of this study was to test the potential interaction between maternal vitamin D status and exposure to ethanol. The main result was that blood alcohol concentration and ADH levels were not affected by Diet. Furthermore, vitamin D levels were assayed and a significant main effect of Diet (Standard vs Deplete) was found, which served to confirm vitamin D deficiency in the DVD group. The blood alcohol concentration assay revealed that while there was an effect of gavage (acute dose used as positive control) versus voluntary drinking, there was no effect of Diet for this measure. Additionally, no Diet x Gavage interaction was found, suggesting that the levels of vitamin D during ethanol exposure did not interfere with the effects induced by ethanol exposure in the offspring.

ADH is one of the major enzyme systems in the liver responsible for oxidation of ethanol (Theorell et al 1961). It was hypothesized that exposure to ethanol would result in increased levels of ADH. However, there was no significant effect of Diet or Gavage on ADH levels. ADH is an enzyme family that, while traditionally associated with ethanol metabolism, has also been shown to participate in retinoid metabolism (important for retinoic acid synthesis from vitamin A) (Duester 1998). Furthermore, ADH has four isoforms (ADH I-IV) with different substrate preference. For example, ADH-IV has been shown to prefer retinol as a substrate over ethanol and ADH-I prefers ethanol. However, it has also been shown that in the presence of excess ethanol, retinol metabolism can be competitively inhibited, whereby ADH isoforms that traditionally prefer retinol would prioritise ethanol metabolism (Duester 1998). The unchanged levels of ADH found in this experiment could be due to the lack of specificity of the assay to detect levels of individual isoforms of ADH and their activity, which may indicate compensatory mechanisms such as the recruitment of added ADH isoforms to oxidise ethanol. Nevertheless, a previous study reported a significant increase in liver weights and in microsomal aniline hydroxylase activity, but no alterations in ADH levels following chronic ethanol administration in C57BL/6J (Kishimoto et al 1995). Liver weights were not measured in this experiment; however, body weight would have also been greater in dams with enlarged liver, and results showed no significant effect of Diet or Exposure in body weight, measured immediately prior to blood and liver tissue collection. Future experiments would need to independently replicate the findings from this experiment, taking into account liver weights and ADH isoforms, although the main finding is consistent with a lack of effect of DVD deficiency on ethanol metabolism.

62

4.3 DVD deficiency and PEE on gene and protein expression

4.3.1 Introduction

Vitamin D is able to regulate gene expression by binding and forming a complex with its receptor (VDR). The complex then heterodimerizes with the retinoid X receptor (RXR), binds to vitamin D response elements (VDREs) located in the promoter region of target genes and in conjunction with coactivators, affects target gene transcription (Christakos et al 2007). This in turn can alter thereby altering molecular processes including cell proliferation and apoptosis (Brown et al 2003, Cui et al 2010, Ko et al 2004, Neveu et al 1994, Wion et al 1991). In rat studies, DVD deficiency has been associated with changes in mitosis, apoptosis, dopamine regulation and gene expression in neonatal brain tissue (Eyles et al 2003). The expression of proteins known to be altered in neuropsychiatric disorders, such as schizophrenia, has also been shown to be dysregulated by DVD deficiency in adult rats (Almeras et al 2007). For example, and neuregulin pathways were altered by DVD deficiency in the neonatal C57BL/6J mouse forebrain (Harms 2011), while brain-derived neurotrophic factor (Bdnf), transforming growth factor- β1 (Tgf-β1), forkhead box protein P2 (FoxP2) and tyrosine hydroxylase (TH) were dysregulated in DVD-deficient BALB/c (Hawes et al 2015), which supports previous findings suggesting that DVD deficiency leads to alterations in the expression of transcription factors important for neurite outgrowth, neuronal survival and dopamine synthesis (Tesic et al 2015).

Another action of vitamin D within the brain is associated with calcium signalling. There is evidence to suggest that the effects of vitamin D on Ca2+ occur via both genomic and non- genomic actions (Eyles et al 2007, Nemere et al 2012, Zanatta et al 2012). Vitamin D can reduce calcium levels by modulating L-type voltage-gated calcium channels (L-VGCCs), which occurs by multiple mechanisms. For example, L-type voltage-sensitive calcium channel (L-VSCC)-A1C subunit mRNA and protein are downregulated via VDR mechanisms (Gezen-Ak et al 2011), while L-VSCC-A1D subunit mRNA can be downregulated with vitamin D treatment, however it is not mediated by VDR (Gezen-Ak et al 2011). Studies in vitamin D-deficient mice have shown upregulation of L-VGCCs, which lead to increased Ca2+ influx (Zhu et al 2012). Moreover, vitamin D has also been shown to regulate the expression of calcium-binding proteins, parvalbumin and calbindin D28k

63

(Dursun et al 2011, Van Cromphaut et al 2001), as well as proteins involved in Ca2+ homeostasis (Eyles et al 2007).

There is evidence to show that prenatal ethanol exposure may disrupt important biological processes, such as increasing cell death (Cartwright et al 1998, Wang et al 2010), reducing proliferation (Jing et al 2004), diminishing glucose metabolism (Singh et al 1989), and disrupting both cell-cell adhesion (Ramanathan et al 1996) and gene transcription. Transcriptional changes have been shown to affect numerous signalling cascades critical for normal development, such as the sonic hedgehog signalling cascade (Ahlgren et al 2002, Li et al 2007, Vangipuram et al 2012), the Wnt/B catenin signalling pathway (Serrano et al 2010, Tong et al 2013, Vangipuram et al 2012) and the Notch signalling pathway (Sathyan et al 2007, Tong et al 2013). Furthermore, ethanol exposure during nervous system development has been shown to disrupt developmental patterns, including neuronal migration (Miller 1986), cell morphology (Davies et al 1981), and ontogeny of neurotransmitter synthesis (Swanson et al 1995). Additionally, PEE has been shown to dysregulate the expression of various neurotrophic factors such as Ngf, Bdnf, neurotrophin-3 and basic fibroblast growth factor (Light et al 2002). However, the majority of these findings have been reported following acute exposure to high doses of ethanol, while the consequences of low-moderate exposure have not been fully described.

The second aim of this chapter was to assess the effects of DVD deficiency and PEE on global changes in gene and protein expression at birth, using hypothesis-generating, unbiased screens on neonatal cerebrum tissue. It was hypothesized that DVD deficiency and PEE would disrupt the expression of molecules relevant to cell death and proliferation, as well as neurotransmitter signalling, and that the combination of DVD and PEE would result in a greater number of affected molecules compared to each single exposure.

4.3.2 Materials and methods

4.3.2.1 Animals, housing and exposures

Twenty adult male and 40 four-week old female C57BL/6J mice were purchased (ARC, Perth, WA) and housed as described in section 2.2.1. The same sires were used throughout the two cohorts of females to reduce variability from different sires. Dams were

64 randomly allocated to one of four experimental groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) following diet and exposure methods described in section 2.2.2 and 2.2.3.

4.3.2.2 RNA-sequencing

a) Neonatal brain tissue collection

A total of 24 samples (n=6/Group) were collected within 12h of birth. Briefly, male and female pups were decapitated and whole cerebrum tissue was dissected. Left and right hemispheres were collected and snap frozen in liquid nitrogen and then stored at -80˚C. Samples were counter-balanced (left and right hemispheres) for RNA-Seq and Proteomics assays.

b) Sample preparation

For preparation of the RNA sequence libraries, 100ng of total RNA was sequenced per sample using the Illumina TruSeq stranded mRNA library LT preparation kit [Illumina, San Diego, CA] according to the manufacturer’s instructions. RNA quality was analysed on the Agilent Bioanalyser and all samples had a RIN number of 8.2 or greater. Samples were also Qubited for RNA input using a HS-RNA assay. 700 ng of RNA was used for library preparation, samples were sequenced in an 8 plex, across 3 lanes of a HiSeq 2000 v4 (50 Gb per lane, 200 million reads per lane minimum).

c) Processing and analysis of RNA-Seq data

Raw reads were aligned to the mouse genome (mm10) using HiSat (Pertea et al 2016). Transcript levels were quantified in HTSeq-count (Anders et al 2015) by mapping to known mouse (mm10) RefSeq (NCBI) protein coding sequences, which were downloaded in a GTF format from the UCSC table browser. In HTSeq-count the overlap mode used was “union”, and the strandedness set to “no”. Differential gene expression analysis was then performed using the DeSeq2 package (Love et al 2014) to compare experimental groups

(DVD-H2O, Std-PEE, DVD-PEE) to control (Std- H2O).

65

4.3.2.3 SWATH Proteomics

a) Sample preparation

A volume of 200ul of extraction buffer (10mM Tris, 8M Urea, 2% SDS) was added to the tissue sample. Tissue pieces were then sonicated on ice (30% amplitude, Vibra-Cell, Sonics, CT, USA). The solution was placed on a rotator and spun slowly for 4 hours at room temperature to complete solubilisation. The samples were then centrifuged at 16000g for 30 minutes at 4°C to remove insoluble debris. The supernatant was removed and placed in a new tube. The sample was then reduced with 10mM Dithiothreitol (DTT) and heated at 30°C in the dry bath incubator for 1 hour. This was followed by alkylation with 25mM idoacetamide (IAA) at room temperature for 15 minutes in the dark. Following alkylation, a 4:1 volume of ice-cold acetone was added to the sample with brief vortexing. The mixture was incubated overnight at -20°C, then centrifuged at 16000g for 30 minutes at 0°C. The pellets were resuspended in 50ul of 10mM Tris, 8M Urea. Once the pellet was completely solubilized, samples were diluted to 2M urea with 50mM ammonium bicarbonate. Protein quantification was performed using a bicinchoninic acid (BCA) assay, according to kit instructions (Pierce, ThermoFisher). The sample was further diluted to 1M urea with 50mM ammonium bicarbonate. Trypsin was added at a 1:50 (enzyme:protein) ratio, and the mixture was incubated overnight at 37°C. Trypsin-digested peptides were purified and concentrated using a ZipTipC18 (10 μL tip with 0.6 μL resin bed, Millipore, Billerica, MA, USA) according to the manufacturer’s instructions. A pooled sample for information-dependent acquisition (IDA) was subjected to strong cation exchange (SCX) fractionation prior to LC-MS/MS.

b) Strong cation exchange fractionation

SCX was performed on an Agilent 1100 series LC system (Agilent Technologies, Mulgrave, Vic, Australia) at a flow rate of 500ul/min. Tryptic peptides were loaded onto a ZORBAX column (Agilent technologies, 50mm x 4.6mm, 5μm) and eluted over 40 min with a linear gradient using ACN buffers (Buffer A: 10mM citric acid, 2% ACN; Buffer B: 10mM ammonium hydroxide, 2% ACN). The elution program was: 10% buffer B for 4 min; 10- 60% for 3 min; 60-97% for 23 min and 10% buffer B for 10 min. Flow-through fractions were collected every 30 seconds (80 in total) into a 96-well plate. Eluate samples # 1-20 were combined to give Fraction 1, #21-24 Fraction 2, #25-28 Fraction 3, #29-32 Fraction

66

4, #33-36 Fraction 5, #37-40 Fraction 6, #41-44 Fraction 7, #45-48 Fraction 8, #49-60 Fraction 9, and #61-80 Fraction 10. Fractions were vacuum concentrated and desalted with the ZipTipC18. Each fraction was processed separately for IDA analysis on the Triple- ToF 5600 mass spectrometer.

c) HPLC Triple-ToF 5600 mass spectrometer

For 1D LC-MS/MS analysis peptides were dissolved in 0.1% formic acid and injected onto an Eksigent nanoLC-Ultra system (AB Sciex) at a flow rate of 300 nL/min onto a cHiPLC- Nanoflex 75 x 150 mm column ChromXP C18-CL 3μm 120 Å phase (Eksigent, USA) configured in trap and elute mode. Chromatographic separation was performed using a linear gradient 1 – 60% solvent B over 90 min. Mobile phases consisted of solvent A (1% acetonitrile, 0.1% formic acid) and solvent B (0.1% formic acid, 90% acetonitrile). The HPLC eluent was interfaced to a TripleTOF 5600 LC/MS/MS system using a Nanospray III ionisation source (Applied Biosystems, Forster City CA). Source conditions included an ion spray voltage of 2700 V, nebulizer gas flow of 10, curtain gas flow of 30, interface heater temperature at 150° C and collision-induced dissociation settings included CAD gas of 10 and a declustering potential of 80 V. MS TOF spectra were acquired across the range m/z350 – 1800 for 0.5 s followed by 20 data-dependent MS/MS measurements on precursor ions (100 counts/s threshold, +2 to +5 charge state, collision energy of 40 with collision energy spread of 15). For SWATH analysis, a second injection was performed in a data independent mode stepped at 25amu across the same mass range. Data was acquired and processed using Analyst TF 1.6™ software.

d) Protein Identification and quantification

ProteinPilot™ v4.5 software (Applied Biosystems, Forster City CA) utilising the Paragon Algorithm was used to analyse the IDA Fractions 1-10. MS/MS data was searched against the Uniprot FASTA formatted database. False discovery rate analysis was done with tools integrated in the software. The ‘thorough’ search setting was selected with the following criteria: identification; cysteine alkylation, iodoacetamide; digestion, trypsin; fixed modification, carbamidomethylation; variable modification, oxidation of methionine; species, mus musculus. The criterion for a positive identification was a confidence score of ≥ 95% and a minimum of 2 peptides per protein.

67

e) Data processing and statistical analysis

The ProteinPilot ion library was imported to the PeakView 2.1 software (AB SCIEX) for quantification of the SWATH samples. Using a 1% False-Discovery Rate (FDR) we were able to import 21619 distinct peptides and 2259 proteins. The number of peptides was set to 30 and transitions to 6. Shared peptides were excluded and modified peptides allowed. The filtered ion library was then exported to the MSstats package using the R platform [26]. The data was subjected to within-group normalisation, to give normalised peak values. Comparisons were made between control (Std-H2O) and the three exposure groups, DVD-H2O Std-PEE and DVD-PEE. Proteins with an adjusted p-value <0.01 were further analysed in PeakView. Each spectral peak was visually checked to confirm its identity. The observed retention times were also checked against the expected retention time. Peptides that could not be confirmed were excluded from the ion library. Proteins with less than two peptides were also removed from the library. The ion library was updated and subjected to statistical reanalysis in MSstats. Proteins with an adjusted p- value <0.01 were considered significant.

4.3.2.4 Pathway analysis

Significant differentially expressed genes and proteins (UniProt) and their fold changes were uploaded to the Ingenuity Pathway Knowledge Base (Ingenuity System Inc, Redwood City, CA). Ingenuity Pathway Analysis was used to identify affected canonical pathways, biological functions and molecular networks. Significance thresholds in these analyses were p<0.01 and fold change of > or < 1.2.

4.3.3 Results

4.3.3.1 RNA-Sequencing

A total of 24 samples (n=6/Group) from neonatal cerebrum tissue were sequenced in a discovery set for gene expression. Analysis using the Fold Change value for the complete list of evaluated molecules, served to highlight the top candidates in each comparison group, for further validation. Furthermore, pathway analysis using Ingenuity Pathway Analysis Tool (Qiagen Bioinformatics) software revealed the top canonical pathways of

68 interest that may be affected, as well as the main molecular and cellular functions that may be affected as a result of such disruptions.

Comparison of each treatment group to controls (Std-H2O) resulted in 57 altered genes in

DVD-H2O, 6 altered genes in the Std-PEE group and 122 altered genes in DVD-PEE samples. Analysis was performed using adjusted p values of altered molecules in every

Group. There was one overlap between DVD- H2O and Std-PEE, and there were no common molecules altered between DVD-PEE and DVD-H2O or Std-PEE (Fig. 4.4). Pathway analyses were performed using IPA ®. (See Appendix 2 for complete list of altered molecular functions and Appendix 3 for list of top dysregulated molecules).

Figure 4.4 Gene expression changes in neonatal brain as a result of DVD deficiency and PEE. Results from whole-genome RNA sequencing study. The Venn diagram depicts the number of genes significantly altered (p<0.05) in each group when compared to controls (Std-H2O).

69

Ingenuity ® Pathway analysis also revealed the top molecular functions affected in each comparison group (Fig. 4.5 A,C,E), along with the number of affected molecules relevant to each function and top dysregulated molecules (based on adjusted p value) were identified (Fig 4.5 B,D,F). DVD deficiency affected 17 molecules involved in molecular transport, 16 transcripts that regulate small molecule biochemistry and 10 molecules that part of cell death and survival signalling. Based on number of altered molecules, PEE resulted in a less extensive effect than DVD deficiency, however, similar molecular functions were identified as vulnerable, including small molecule biochemistry (3 altered molecules), cell-to-cell signalling and interaction (1), and cell death and survival (1) identified as vulnerable. The combination of factors (DVD-PEE) resulted in the largest number of dysregulated genes, whereby 23 of the altered molecules are involved in cellular assembly and organization, 21 regulate cellular function and maintenance, and 15 play a role in small molecule biochemistry. IPA ® was also used to generate network analysis for each comparison group, where up and downregulated molecules were arranged based on cellular localization and potential regulatory pathways (Fig. 4.6-4.8).

70

Std-H2O vs DVD-H2O A B Molecular Transport

Cell Death & Survival (4.9E-02 - 1.3E-03) KCNQ5 PTGDS

Drug Metabolism (2.1E-02 - 3.1E-03) Cell Death & Survival

NECAB1 CRABP2 Amino Acid Metabolism (1.8E-02 - 3.1E-05)

Small Molecule Biochemistry (4.9E-02 - 2.5E-06)

Molecular Transport (4.9E-02 - 2.5E-06)

0 5 10 15 20 25 No. altered molecules

Std-H2O vs Std-PEE C D Small Molecule Biochemistry

C4A/C4B. FGFBP3 Cell Death & Survival (6.8E-03 - 2.1E-03)

Cell-To-Cell Signaling and Interaction (3.6E-02 - 1.9E-03) Molecular Transport

Small Molecule Biochemistry (1.3E-02 - 9.4E-04) Ubb RPL23

Molecular Transport (1.3E-02 - 9.4E-04)

Amino Acid Metabolism (1.3E-02 - 9.4E-04)

0 5 10 15 20 25 No. altered molecules

Std-H2O vs DVD-PEE E F Cellular Assembly & Organization

Cellular Function & Maintenance (3.2E-02 - 4.9E-04) NR1D1 F13A1 Cellular Assembly and Organization (2.8E-02 - 4.8E-04) Cellular Function Small Molecule Biochemistry (3.2E-02 - 6.4E-05) & Maintenance

Nucleic Acid Metabolism (2.3E-02 - 6.4E-05) PER3 DENND1C

Cell Signaling (1.4E-02 - 6.4E-05)

0 5 10 15 20 25 No. altered molecules

Figure 4.5 RNA-Sequencing results from neonatal brain tissue Molecular functions altered as a result of (A) DVD deficiency, (C) PEE and (E) DVD-PEE ranked according to number of altered molecules relevant to each function. –log10 p-value range is represented in parentheses. (B, D, E) Curated lists of genes associated with altered molecular functions in each comparison group. (n=6/Group)

71

Further analysis revealed the top canonical pathways disrupted in each comparison group (Table 4.1). Some of these findings served to confirm previous reports found in the literature, such as the effects of DVD deficiency on synaptic long-term potentiation (p=2.68E-03), DA-DARPP-32 feedback in cAMP signalling (p=6.20E-03) and Wnt/Ca+ pathway (p=7.67E-03). Other findings were not predicted, such as the impact of PEE on complement system (p=8.68E-03), or the strong dysregulation of circadian rhythm signalling (p=4.84E-04) as a result of DVD-PEE.

Table 4.1 List of top canonical pathways altered by DVD deficiency and PEE on gene expression.

Top Canonical Pathways Std-H2O v. DVD-H2O Name p-value Overlap Synaptic Long-Term Potentiation 2.68E-03 2.5 % 3/120 Dopamine-DARPP32 Feedback in cAMP Signalling 6.20E-03 1.9 % 3/162 Wnt/Ca+ pathway 7.67E-03 3.5 % 2/57 PRPP Biosynthesis I 9.19E-03 25.0 % 1/4 Glycerol-3-phosphate Shuttle 9.19E-03 25.0 % 1/4

Std-H2O v. Std-PEE Complement System 8.68E-03 2.7 % 1/37 GABA Receptor Signalling 1.57E-02 1.5 % 1/67 Toll-like Receptor Signalling 1.73E-02 1.4 % 1/74 Renal Cell Carcinoma Signalling 1.89E-02 1.2 % 1/81 LXR/RXR Activation 2.82E-02 0.8 % 1/121

Std-H2O v. DVD-PEE Circadian Rhythm Signalling 4.84E-04 9.1 % 3/33 Granzyme A Signalling 3.87E-03 10.0 % 2/20 -alanine Degradation I 9.30E-03 50.0 % 1/2 Inhibition of Matrix Metalloproteases 1.42E-02 5.1 % 2/39 Neuroprotective Role of THOP1 in Alzheimer's Disease 1.49E-02 5.0 % 2/40

72

Figure 4.6 IPA® network analysis of dysregulated molecules in DVD-H2O samples. Molecules found to be disrupted by DVD deficiency, organized into networks and localization (nucleus, cytoplasm, plasma membrane, extracellular space), showing possible pathway interactions. Molecules in green were downregulated, while those in red were upregulated. Molecules in grey were not significantly altered but are important downstream/upstream members of dysregulated pathways.

73

Figure 4.7 IPA® network analysis of dysregulated molecules in Std-PEE samples. Molecules found to be disrupted by PEE, organized into networks and localization (nucleus, cytoplasm, plasma membrane, extracellular space), showing possible pathway interactions. Molecules in green were downregulated, while those in red were upregulated. Molecules in grey were not significantly altered but are important downstream/upstream members of dysregulated pathways.

74

Figure 4.8 IPA® network analysis of dysregulated molecules in DVD-PEE samples. Molecules found to be disrupted by the combination of DVD deficiency and PEE, organized into networks and localization (nucleus, cytoplasm, plasma membrane, extracellular space), showing possible pathway interactions. Molecules in green were downregulated, while those in red were upregulated. Molecules in grey were not significantly altered but are important downstream/upstream members of dysregulated pathways.

75

4.3.3.2 SWATH Proteomics

The opposite hemisphere to that of samples used for RNA-Seq was prepped and assayed in a proteomics screen (n=6/Group). Results were analysed with IPA ®. Similar to RNA-

Seq analysis, each treatment group was compared to controls (Std-H2O). Top molecules (based on adjusted p value) were matched to test for overlapping changes (Fig. 4.9).

There were 12 altered molecules in the DVD-H2O group, 13 in the Std-PEE group and 11 in the DVD-PEE group. Furthermore, there were 2 common affected molecules between

DVD-H2O and Std-PEE; 4 between DVD-H2O and DVD-PEE; 3 between Std-PEE and DVD-PEE; and 2 molecules were present in all comparison groups. (See Appendix 4 for complete list of altered molecular functions and Appendix 5 for list of top dysregulated molecules).

Figure 4.9 Protein expression changes in neonatal brain as a result of DVD deficiency and PEE. Results from cerebrum Proteomics study. The Venn diagram depicts the number of proteins significantly altered (p<0.05) in each group when compared to controls (Std-H2O). n=6/Group.

76

Top altered canonical pathways and corresponding molecular and cellular functions were also identified. Some of the most relevant molecular functions found to be affected in DVD- deficient samples, at protein level, were cell death and survival and cell function and maintenance, with 21 and 17 dysregulated proteins, respectively (Fig. 4.10 A). In PEE samples, cell assembly and organization, as well as cell function and maintenance were found to be disrupted, which involved 18 and 16 dysregulated proteins, respectively (Fig. 4.10 C). The combination of exposures (DVD-PEE) did not result in an exacerbated effect of molecular functions disrupted by a single hit (DVD or PEE). Cell death and survival was altered, consistent with findings form DVD-deficient samples; however, there were 10 proteins dysregulated in this pathway, as opposed to 21 found in the DVD deficiency group. In agreement with changes in PEE samples, cell morphology was dysregulated in the combination group, although the number of affected molecules part of this function was not aggravated as a result of the combination of factors (15 affected molecules in PEE group, 13 in DVD-PEE) (Fig 4.10 E).

77

A Std-H2O vs DVD-H2O B Molecular Transport

ETFA Molecular Transport (3.1E-02 - 6.7E-04) Protein Synthesis Cellular Function & Maintenance (3.4E-02 - 4.4E-04) GNA11 Protein Synthesis (3.1E-02 - 2.7E-04) Cell Death & Survival

(3.4E-02 - 1.2E-04) Cell Death & Survival COTL1 RPL7

RNA Post-Transcriptional Modification (2.01E-02 - 1.02E-05)

0 5 10 15 20 25 No. altered molecules

C Std-H2O vs Std-PEE D Cellular Assembly & Organization

(1.7E-02 - 1.4E-06) Cellular Growth & Proliferation NUDT21. SCAMP1

Cellular Function & Maintenance (2.2E-02 - 1.4E-06) Cellular Growth & Proliferation (1.7E-02 - 1.4E-06) Cellular Development

Cellular Assembly & Organization (1.9E-02 - 1.4E-06) GPC2 Cellular Function Cell Morphology (1.9E-02 - 1.4E-06) & Maintenance

0 5 10 15 20 25 MARS No. altered molecules

E Std-H2O vs DVD-PEE F Cell Morphology

LARP1 Cell Death and Survival (4.9E-02 - 9.7E-04) Cell Death Cell Morphology (4.9E-02 - 7.4E-04) & Survival

Protein Synthesis (4.3E-02 - 5.4E-04) COTL1

Carbohydrate Metabolism (3.6E-02 - 2.2E-04) Protein Synthesis

RNA Post-Transcriptional Modification (1.8E-02 - 5.3E-05) PSMD13 PSMA5

0 5 10 15 20 25 No. altered molecules

Figure 4.10 SWATH Proteomics results from neonatal brain tissue. Molecular functions altered as a result of (A) DVD deficiency, (C) PEE and (E) DVD-PEE ranked according to number of altered molecules relevant to each function. –log10 p-value range is represented brackets. (B, D, E) Curated lists of proteins associated with altered molecular functions in each comparison group (n=6/Group).

78

The canonical pathways (Table 4.2) with the largest effect in the DVD-deficient group were DNA damage checkpoint regulation (p=3.85E-04) and TCA cycle II (p=2.00E-03). In PEE samples, remodelling of epithelial adherens junctions (p=4.39E-04) and mitochondrial dysfunction (p=6.14E-03) were the most vulnerable pathways. The combination of DVD- PEE resulted in dysregulated tight junction signalling (p=3.60E-03) and methylglyoxal degradation I (p=5.53E-03). These are exclusive pathways from those altered as a result of a single exposure, suggesting DVD deficiency does not exacerbate the effects of PEE. Additionally, the changes in canonical pathways affected by DVD-PEE had a lower level of significance compared to dysregulations observed in DVD or PEE alone.

Table 4.2 List of top canonical pathways altered by DVD deficiency and PEE on protein expression.

Top Canonical Pathways Std-H2O v. DVD-H2O Name p-value Overlap Cell Cycle: G2/M DNA Damage Checkpoint Regulation 3.85E-04 6.1 % 3/49 TCA Cycle II (Eukaryotic) 2.00E-03 8.7 % 2/23 PRPP Biosynthesis I 1.15E-02 25.0 % 1/4 Tight Junction Signalling 1.25E-02 1.8 % 3/167 RhoGDI Signalling 1.37E-02 1.7 % 3/173

Std-H2O v. Std-PEE Remodelling of Epithelial Adherens Junctions 4.39E-04 4.4 % 3/68 Mitochondrial Dysfunction 6.14E-03 1.8 % 3/171 CMP-N-acetylneuraminate Biosynthesis I (Eukaryotes) 1.08E-02 20.0 % 1/5 Oxidative Phosphorylation 2.36E-02 1.8 % 2/109 Cleavage and Polyadenylation of Pre-mRNA 2.58E-02 8.3 % 1/12

Std-H2O v. DVD-PEE Tight Junction Signalling 3.60E-03 1.8 % 3/167 Methylglyoxal Degradation I 5.53E-03 33.3 % 1/3 Arsenate Detoxification I (Glutaredoxin) 7.37E-03 25.0 % 1/4 PRPP Biosynthesis I 7.37E-03 25.0 % 1/4 Ascorbate Recycling (Cytosolic) 7.37E-03 25.0 % 1/4

79

4.3.4 Discussion

The main aim of the second study presented in this chapter was to undertake two broad screens to reveal possible candidate pathways and molecules affected as a result of DVD, PEE and their combination at transcript and protein level. The first approach consisted of RNA-sequencing, using RNA extracted from neonatal cerebrum tissue, to highlight individual candidates for future studies, and to identify molecular functions and canonical pathways vulnerable to each exposure and their combination. The second approach examined protein levels using SWATH proteomics on neonatal cerebrum tissue, as well as their corresponding molecular functions and canonical pathways. Both of these approaches should be considered preliminary, with the intention of identifying candidates of large effect expressed throughout the whole brain. The findings from the RNA-seq data support the hypothesis that the effects of DVD deficiency are exacerbated by the addition of a secondary hit, in this case PEE, whereas the findings from the SWATH data did not. These results are discussed in more detail in the sections below.

4.3.4.1 Effects of DVD deficiency and PEE on neonatal gene expression

The top canonical pathways and affected molecular functions in each comparison were identified from RNA-Seq analysis. Those listed as dysregulated in the DVD-H2O and Std- PEE groups were not novel, however, they served to corroborate findings previously reported in the literature. Nevertheless, this is the first study to examine the effects of the combination of DVD deficiency and PEE on gene expression.

The comparison of control samples (Std-H2O) vs. DVD-H2O highlighted Synaptic Long- Term Potentiation, Dopamine-DARPP32 Feedback in cAMP Signalling and Wnt/Ca+ signalling as the main disrupted pathways, which in turn affect important molecular functions such as molecular transport, small molecule biochemistry and cell death and survival. Previous DVD studies have shown that the absence of vitamin D impacts on cell, growth, survival and morphology. Apoptosis and cell cycle targeted cDNA expression arrays on cortical rat tissue have shown reduced expression of pro-apoptotic genes, such as Bak, at E19 and P0, increased expression of pro-mitotic genes, including cyclins A1, D1 and E and decreased levels of anti-mitotic gene, p21 (Ko et al 2004). Moreover, qRT-PCR analyses on midbrain tissue from DVD-deficient embryos have shown decreased expression of a nuclear receptor essential for the development and maturation of dopaminergic neurons, NURR1 (Cui et al 2010). The mRNA levels of COMT, the enzyme

80 that catalyses the conversion of DOPAC to HVA, were also reduced in DVD-deficient rat tissue, suggesting DVD deficiency may have an impact on dopaminergic development (Kesby et al 2009).

Comparing Std-H2O vs. Std-PEE revealed that the complement system, GABA receptor signalling, and LXR/RXR activation were the top dysregulated pathways, while the affected molecular functions included amino acid metabolism, cell-to-cell signalling and interaction and cell death and survival. Some of these findings were not predicted, such as the impact of PEE on the complement system. Taking into consideration novel information recently published regarding C4 regulation and risk for neurodevelopmental disorders (see C4A/C4B discussion above), these results suggest that the effects of PEE on the regulation of the complement system may have a significant impact on the risk for neurodevelopmental disorders.

Other studies on PEE have shown that ethanol has a varying degree of impact on gene expression, based on dose and timing of exposure. Using the same model of moderate PEE as this study, transcriptional changes were found in the embryo (E9), gene and miRNA expression changes were identified in the hippocampus of adolescent (P28) and adult mice (P80). Unbiased screens of mRNA and miRNA expression at E9, only identified one gene (Bcl11b) out of ∼45, 000 probes on the microarray (annotated to 30, 855 genes) and 641 miRNAs to be altered as a result of PEE (Yamada 2017). A genome-wide analysis reported 23 genes and 3 miRNAs as altered in the hippocampus of PEE adolescent offspring, and DNA methylation in the CpG islands upstream of the candidate genes was also altered (Marjonen et al 2015). Furthermore, another study reported a disruption of the developmental silencing of Slc17a6, which encodes for the vesicular glutamate transporter Vglut2, in the hippocampus of adult PEE offspring (Zhang et al 2015). These preliminary findings suggest that even a short, moderate dose can alter gene expression regulation, which may be associated with long-term phenotypic changes.

Further investigation revealed individual molecules altered in each comparison group.

While there were 57 transcripts affected in the DVD-H2O group, there were only 6 altered genes in the Std-PEE group and the DVD-PEE showed the largest number of affected molecules (122). It is important to note that overlapping molecules (altered in two or more comparisons) were minimal, only one candidate was present in both DVD-H2O and Std- PEE, Crystaline Mu (Crym), a protein-coding gene that binds NADPH; its encoded protein

81 binds thyroid hormone and it may have developmental or regulatory roles. Crym mutations have been associated with autosomal dominant non-syndromic deafness. A study on human and mouse foetal cerebral cortices exposed to ethanol in vitro and in vivo, respectively, reported a significant downregulation of Crym compared to controls (Hashimoto-Torii et al 2011). Furthermore, there were no common changes between DVD- PEE and the single –hit comparisons, which suggests that when combined, these exposures dysregulate different molecules and pathways, independent form those affected by single-hit exposures.

The top upregulated molecules in the DVD deficiency group were KCNQ5 and NECAB1, while PTGDS and CRABP2 were the top downregulated transcripts. Potassium Voltage- Gated Channel Subfamily Q Member 5 (KCNQ5), is a potassium voltage-gated channel, the most recently discovered member of the KCNQ family of potassium channel genes. It is differentially expressed in subregions of the brain and in skeletal muscle. KCNQ5 was found to be widely distributed throughout the temporal neocortex and the hippocampal formation (Yus-nájera et al 2003). In these structures, both pyramidal and non-pyramidal neurons and a population of glial cells in the white matter expressed the KCNQ5 subunit. Furthermore, there is more extensive literature on other members of the KCNQ family. Mutations in KCNQ2 and 3 results in neonatal generalized epilepsy; these form heteromeric potassium channels expressed broadly throughout the brain and the associated current is almost identical to the M-current, which is an important regulator of neuronal excitability (Wang et al 2016a). A more recent study reported that KCNQ5 largely overlaps with KCNQ2 and 3 and can form hetermeric channels with KCNQ3. Currents expressed from KCNQ5 have voltage dependences and inhibitor sensitivities in common with M-currents, indicating a molecular diversity of channels yielding M-type currents and suggests KCNQ5 may have a role in the regulation of neuronal excitability (Schroeder et al 2000). Another study in control and epileptic patients reported a marked loss of KCNQ5 immunoreactive pyramidal neurons in the sclerotic areas of the CA fields of epileptic patients, suggesting KCNQ5 may also be involved in neonatal generalized epilepsy (Yus- nájera et al 2003). Furthermore, there is some evidence to show an association between vitamin D and epilepsy. Epidemiological studies have reported a seasonal pattern of epilepsy in newborns, where the rate of epileptic births was highest in the winter months (Procopio et al 1998, Procopio et al 2006, Procopio et al 1997). Meanwhile, studies in animals have shown that administration of vitamin D can significantly increase seizure threshold, decrease the severity of chemically induced seizures, and enhance the

82 anticonvulsant effect of phenytoin and valproate (Borowicz et al 2007, Kalueff et al 2005, Siegel et al 1984). Moreover, VDR-knockout rats showed increased seizure susceptibility (Kalueff et al 2006a, Kalueff et al 2006b). The mechanism by which vitamin D deficiency may increase the risk for epilepsy is not fully understood, though one possible explanation is considering that vitamin D plays part in the regulation of voltage-gated calcium and potassium channels, its absence may result in an imbalance between excitatory and inhibitory currents that may lead to epileptic seizures.

There is limited information on the function of N-terminal EF-hand calcium binding protein 1 (NECAB1) in the brain. However, it is part of the neuronal calcium-binding family and is expressed in layer 4 pyramidal neurons of the , and in inhibitory interneurons and CA2 pyramidal cells of the hippocampus in rats (Sugita et al 2002). Ca2+ plays a crucial role in many and diverse cellular processes, including neurotransmission.

Neurotransmitter release is tightly regulated by Ca2+ -dependent SNARE proteins whose activity is regulated by Ca2+ -binding proteins (CaBPs). A high level of calcium in the brain leads to neurotoxicity, and one of the neuroprotective qualities of vitamin D is to promote a reduction in calcium levels. Vitamin D has been shown to downregulate L-VGCCs (Zanatta et al 2012, Zhu et al 2012), as well as to regulate the gene expression of other calcium- binding proteins, including parvalbumin and calbindin D28k, and proteins associated with

Ca2+ homeostasis (Dursun et al 2011, Eyles et al 2007, Van Cromphaut et al 2001). The upregulation of NECAB1 upon the absence of vitamin D has not been previously described, however it is consistent with findings highlighting the neuroprotective actions of vitamin D.

PTGDS was the top downregulated molecule in the DVD group. The protein encoded by this gene is a glutathione-independent prostaglandin D synthase, which catalyses the conversion of prostaglandin H2 (PGH2) to prostaglandin D2 (PGD2) (Igarashi et al 1992). PGD2 is a neuromodulator as well as a trophic factor in the central nervous system. At cellular level, it is involved in apoptosis, migration and differentiation, and its dysregulation has been associated with various types of cancer, including glioblastoma, prostate and lung cancer (Yoshida et al 1998). Vitamin D has been previously shown to regulate the expression of apoptotic genes, which makes PTGDS another plausible candidate as a downstream target of vitamin D regulation.

83

Cellular retinoic acid binding protein 2 (CRABP2) was also downregulated as a result of DVD deficiency. It encodes a member of the retinoic acid (RA) protein family. The protein is a cytosol-to-nuclear shuttling protein, involved in the retinoid signalling pathway by facilitating RA binding to its receptor complex and transfer to the nucleus (Astrom et al 1991). Studies in neuroblastoma cells have reported that CRABP2 mRNA expression can be inhibited in the presence of a retinoid X receptor (RXR)-homodimer antagonist (Hewson et al 2002). Moreover, VDR has been shown to heterodimerise with RXR and to bind target DNA sequences to regulate gene transcription (Barsony et al 2002), making CRABP2 a plausible downstream target affected by DVD deficiency.

With regards to the Std-PEE group, the top upregulated molecules were C4A/C4B and Ubb, while FGFBP3 and RPL23 were the top downregulated transcripts. C4A/C4B encodes the basic form of complement factor 4, it is a critical component of the classical complement cascade, an innate pathway that rapidly recognizes and eliminates pathogens and cellular debris. In the brain, C4+ cells have been observed in grey and white matter, with the greatest number of C4+ cells found in the hippocampus. Additionally, C4 has also been detected in subsets of NeuN+ neurons and a subset of astrocytes, suggesting it is produced by, or deposited on, neurons and synapses (Sekar et al 2016). In mice, C4 has been shown to promote synapse elimination during the developmentally timed maturation of neuronal circuits. Moreover, the role of C4 in synaptic pruning has been recent explored in relation to schizophrenia risk, where post-mortem studies revealed that the median expression of C4A in brain tissues from schizophrenia patients was 1.4-fold greater than in healthy controls (Sekar et al 2016). This suggests that there may be excessive complement activity in the development of schizophrenia, and provides a possible explanation for the reduced numbers of synapses in the brains of schizophrenia patients. C4 has also been associated with other neurodevelopmental disorders, including autism and ADHD, whereby one study found plasma concentration of the C4B protein in autistic patients was significantly lower than that of control subjects (Warren et al 1994) and another study reported C4B plasma levels in ADHD subjects to be significantly decreased compared to those in the control age-matched subjects (Warren et al 1995). Furthermore, considering PEE has been previously associated with increasing the risk for neurodevelopmental disorders in the offspring, C4A/C4B is an important candidate which, upon validation, would provide valuable insight into the mechanism underlying the teratogenic effects of ethanol.

84

Ubb was significantly upregulated in the Std-PEE comparison group. It encodes ubiquitin, a highly conserved protein that has a major role in targeting cellular proteins for degradation. It is also involved in the maintenance of chromatin structure, the regulation of gene expression, and stress response. Disruption of Ubb leads to early-onset reactive gliosis and adult-onset hypothalamic neurodegeneration in mice (Ryu et al 2014), though the underlying mechanisms are not fully understood. A study in vitro, using isolated cells from Ubb (-/-) mouse embryonic brain tissue identified an unexpected role for cellular Ub pools in determining the fate and self-renewal of neural stem cells (NSCs). The study reported an increase in number of glial cells and decreased number of neurons; moreover, reduced proliferation and premature differentiation into glial cells was observed, which resulted in a significant decrease in the number of neural stem cells. Additionally, Ubb (-/-) cells showed a significant reduction of pro-neuronal gene expression, possibly mediated by increased steady-state levels of Notch intracellular domain, which leads to an upregulation of Notch target genes (Ryu et al 2014). Furthermore, PEE studies in vitro have shown that numerous genes involved in the ubiquitin pathway, protein synthesis and chromatin remodelling and methylation are dysregulated upon ethanol exposure (Gutala et al 2004, Liu et al 2009, Zhou et al 2011) (Downing et al 2012, Mason et al 2012), suggesting these are important targets of PEE.

Fibroblast growth factor binding protein 3 (FGFBP3) was the top downregulated molecule in PEE samples. Clinical studies have shown that FGF signalling is disrupted in patients with major depressive disorders (Evans et al 2004, Gaughran et al 2006), and in mice FGF-2 expression levels were decreased after prenatal stress or social defeat (Turner et al 2008). Moreover, FGFBP3 has been shown to bind to FGF-2 in vitro, and to colocalize with FGF-2 in vivo, thereby suggesting FGFBP3 may be important in the stabilization of FGF-2 by acting as a molecular chaperone (Yamanaka et al 2011). Furthermore, a study using mice with a targeted deletion of FGFBP3 found that its inactivation resulted in altered anxiety-related behaviours, including reduced time spent in the centre of an open field arena, reduced activity in lit areas of light/dark transition box, as well as prolonged latency to feed during a novelty-induced hypophagia test. These behavioural findings were correlated with alterations in light-induced orbitofrontal cortex (OFC) activation via extracellular signal-regulated kinase (ERK) pathway-dependent manner (Yamanaka et al 2011). There is no direct evidence showing that prenatal ethanol disrupts FGFBP3; however, in-vitro studies have shown that ethanol can inhibit basic FGF-mediated

85 proliferation of glial and neuronal precursors in a dose-dependent manner (Luo et al 1996), which suggests that FGFBP3 may also be a plausible candidate vulnerable to PEE.

Ribosomal protein L23 (RPL23) was also significantly downregulated in the PEE group. RPs, which are components of ribosomal subunits, are ubiquitous RNA-binding proteins that carry out multiple extraribosomal functions. RPL23 is a protein component of the 60S large ribosomal subunit, which has been shown to negatively regulate apoptosis (Qi et al 2017). When ribosome biogenesis is dysregulated, ribosomal proteins, including RPL23, interact with MDM2, an E3 ubiquitin ligase, to suppress its ubiquitinase activity and thereby stabilize the tumour suppressor protein and stimulate p53-mediated activities including cell cycle arrest and apoptosis (Fumagalli et al 2011). Ribosomopathies in humans are not embryolethal, however, they are associated with significant craniofacial deficits such as flattened nasal bridge, micrognathia, epicanthal folds, cleft lip/palate, and altered palpebral fissues (Fumagalli et al 2011, Narla et al 2010), which are also commonly observed in FASD cases, thereby suggesting that cranial neural crest is particularly sensitive to altered ribosome biogenesis (Smith et al 2014).

In the DVD-PEE group, the top upregulated molecules were NR1D1 and PER3, while F13A1 and DENND1C were the top downregulated molecules. Nuclear receptors are an evolutionarily conserved group of transcription factors that regulate genes involved in multiple functions such as homeostasis, development, metabolism and immune response. Nuclear receptors bind to lipophilic-ligands such as steroid hormones, including vitamin D, to modulate transcriptional activity (Fuller 1991). NR1D1 appears to have a diverse role in regulating gene networks in many tissue types and several biological processes. It is able to function as a monomer or a dimer, and is capable of repressing or activating gene expression (Adelmant et al 1996, Harding et al 1993, Harding et al 1995). NR1D1 has been widely studied for its role in transcriptional regulation of processes such as circadian rhythm and metabolism, with heme functioning as an endogenous ligand (Raghuram et al 2007, Yin et al 2010). Evidence correlating NR1D1 and PEE is lacking; a genetic screen study in adolescents in relation to alcohol use disorders (AUD) found an association with circadian rhythm genes, one of them was NR1D1, which suggests that the circadian pathway may be relevant to ethanol pathophysiology (Dalvie et al 2016).

Period circadian protein homolog 3 (PER3) was the second most upregulated molecule in DVD-PEE brain samples. It is a member of the Period family of genes and it is expressed

86 in a circadian pattern in the suprachiasmatic nucleus, the primary circadian pacemaker in the mammalian brain. Genes in the Period family encode components of the circadian rhythms of locomotor activity, metabolism, and behaviour. Polymorphisms in PER3 have been linked to sleep disorders (Dallaspezia et al 2016, Hong et al 2015). Per3 basal expression has also been correlated with anxiety-like and addiction-related phenotypes in rats. A study in mice reported that PEE resulted in increased expression of Per3 in the hippocampus, which was correlated to acute restraint stress response (Wang et al 2012). Another study in rats found that PEE affects circadian function of the stress-axis regulatory -endorphin neurons in the hypothalamus of adult offspring, as well as the circadian expression of Per1, Per 2 and Per3 in the arcuate nucleus (Chen et al 2006). This provides further evidence to suggest that ethanol exposure is associated with the regulation of Period genes and is able to alter the mechanisms governing circadian function.

F13A1, or coagulation factor XIII A chain, was the top downregulated molecule in the DVD-PEE group. Coagulation factor XIII is part of the blood coagulation cascade. Plasma factor XIII is a heterotetramer composed of 2 A subunits and 2 B subunits. The A subunits have catalytic function, and the B subunits do not have enzymatic activity and may serve as plasma carrier molecules. Platelet factor XIII is comprised only of 2 A subunits, which are identical to those of plasma origin. Upon cleavage of the activation peptide by thrombin and in the presence of calcium ion, the plasma factor XIII dissociates its B subunits and yields the same active enzyme, factor XIIIa, as platelet factor XIII. This enzyme acts as a transglutaminase to catalyse the formation of gamma-glutamyl-epsilon- lysine crosslinking between fibrin molecules, which stabilize the fibrin clot. F13A1 has been previously studied in reference to neurodegenerative disorders, such as Alzheimer’s Disease (AD). A study using iTRAQ-based LC-MS/MS analyses of controls with negative PiB-PET scores and mild cognitive impairment (MCI) and AD with positive PiB-PET scores reported and validated 3 biomarker candidates, F13A1, PCSK9 and DCD, as upregulated proteins in both MCI and AD, compared to controls (Kang et al 2016). F13A1 was also immunohistochemically detected in reactive microglia during gliosis, which is a hallmark feature of AD pathogenesis (Gerardino et al 2006). Additionally, a common polymorphism of F13A1 has been previously associated with sporadic AD (Gerardino et al 2006).

DENN domain containing 1C (DENND1C) encodes a protein that functions as a guanine nucleotide exchange factor for the early endosomal small GTPase RAB35, which

87 regulates endosomal membrane trafficking and is involved in actin polymerization. The encoded protein activates RAB35 by promoting the exchange of RAB35-bound GDP for GTP. This gene may play a role in linking RAB35 activation with the clathrin machinery. There is little evidence on the effects of DVD deficiency or PEE on the expression of DENND1C. However, a study on ethanol exposure in rats reported upregulation of RAB35 in ethanol-administering rats compared to controls (Bell et al 2009); considering DENND1C is involved in RAB35 activation, it is possible that the PEE may alter RAB35 regulation via disruptions in DENND1C expression levels.

4.3.4.2 Effects of DVD deficiency and PEE on neonatal protein expression

Unbiased SWATH proteomics analysis identified the top altered molecules in each comparison group. The findings from this assay, while preliminary, served to test the biological plausibility of the effects of DVD deficiency and PEE on the regulation of cellular processes important for neurodevelopment. Pathways and molecular functions affected included Molecular Transport, Cell Death and Survival, Cellular Function and Maintenance and Cell Death and Survival, which are somewhat consistent with processes highlighted during RNA Sequencing assessment. However, pathway analysis comparing results from RNA sequencing and proteomics assays showed that while there was a large number of molecules listed as affected at transcript (RNA) level, there was minimal translation of these alterations resulting in dysregulated proteins, which is a common finding due to the high number of regulatory steps that occur between gene and protein expression (Ghazalpour et al 2011).

Electron Transfer Flavoprotein Alpha Subunit (ETFA) and Coactosin-like protein (COTL1) were the most significantly dysregulated candidates as a result of DVD deficiency. ETFA, which was also altered in DVD-PEE samples, serves as a specific electron acceptor for several dehydrogenases; it transfers the electrons to the main mitochondrial respiratory chain via ETF-ubiquinone oxidoreductase. There is no previous association of vitamin D with this protein, however, in-vitro assays have shown that genetic silencing of the VDR results in potentiated mitochondrial respiratory chain protein levels (Consiglio et al 2015). In addition, several studies have reported a strong association between mitochondrial abnormalities and ASD in children, and have postulated that mitochondrial dysfunction

88 may be critical in the causal pathway that determines abnormalities in brain function and structure in autism (Siddiqui et al 2016), thereby suggesting that the role of DVD deficiency in the regulation of ETFA may have an impact on the development of disorders such as ASD. COTL1 is an actin binding and enzyme binding protein; it binds to F-actin in a calcium-independent manner and acts as a chaperone for ALOX5, influencing both its stability and activity in leukotrienes synthesis. COTL1 was found to be dysregulated in all comparison groups, however, there have not been previous associations between COTL1 and DVD deficiency or PEE in the literature.

G protein subunit alpha 11 (GNA11) and ribosomal protein L7 (RPL7) were the top downregulated molecules in the DVD deficiency group. GNA11 is part of the protein family of guanine nucleotide-binding proteins, or G proteins. It is a key mediator of Ca2+ sensing receptor (CASR) signalling, and dysregulation of this molecule has been associated with disorders such as hypocalciuric hypercalcaemia type II and hypocalcaemia dominant 2, where patients present impaired sensitivity to changes in extracellular calcium concentrations (Nesbit et al 2013). With regards to vitamin D, previous studies have observed a complex cross-talk between vitamin D and CASR, vitamin D-depleted rats had significantly reduced CASR expression (Brown et al 1996, Hendy et al 2016), and administration of 1,25(OH)D3 upregulated CASR expression in various tissues (Canaff et al 2002). This evidence suggests that the regulatory actions of vitamin D (or absence thereof) on GNA11 may be a plausible mode of action by which vitamin D is involved in

Ca2+ signalling. RPL7 is a component of the large ribosomal subunit, which plays a regulatory role in the translation apparatus and inhibits cell-free translation of mRNA (Hemmerich et al 1993). This protein interacts with the VDR in the presence of vitamin D and has been shown to be a core regulator of VDR-RXR mediated transactivation of genes (Berghofer-Hochheimer et al 1998), which suggests it is a plausible candidate involved in the role of vitamin D in gene expression regulation.

In the Std-PEE group, polyadenylation specificity factor subunit 5 (NUDT21) and secretory carrier membrane protein 1 (SCAMP1) were the top upregulated molecules, while glypican 2 (GPC2) and methionine-tRNA ligase (MARS) were the top downregulated proteins. NUDT21 is part of the nudix superfamily of superfamily of hydrolytic enzymes capable of cleaving nucleoside diphosphates linked to x (any moiety). It is also a component of the cleavage factor Im (CFIm) complex that plays a key role in pre-mRNA 3’ processing (Ruegsegger et al 1998). This protein was also altered in DVD and DVD-PEE samples;

89 however, there is limited information regarding the relationship between vitamin D or ethanol and NUDT21. PEE gene expression analyses have identified other proteins from the nudix family, such as NUDT3, 8 and 22 to be altered as a result of exposure to ethanol (Camargo Moreno et al 2017, Kleiber et al 2013, Mulligan et al 2008), however the differing dose, timing of exposure and of tissue collection could account for the absence of alterations in NUDT21 in these studies. SCAMP1 belongs to the SCAMP family of proteins, which function as carriers to the cell surface in post-golgi recycling pathways. An association between SCAMP1 and PEE has not been previously reported. Chronic alcohol exposure studies have reported that ethanol alters the lipidation of Rab proteins, which leads to the inhibition of protein trafficking at the ER and Golgi apparatus (Nagy et al 2002), perhaps ethanol exposure also has a role in the regulation of other secretory membrane proteins, including SCAMP1.

GPC2, also known as cerebroglycan, is exclusively expressed in the developing nervous system in a transient pattern. It appears to participate in cell adhesion and regulation of cell growth and guidance (Stipp et al 1994). Studies in rats have shown prominent GPC2 expression on axon tracts while were actively growing, but generally not after axons had reached their targets (Ivins et al 1997). Indeed, GPC2 ceased to be expressed in mature neurons, and it was only present in newly generated granule neurons of the hippocampus at a late stage in development (Ivins et al 1997). Although a direct association between PEE and GPC2 has not been previously described, there is vast evidence in the literature regarding the effects of ethanol on cell adhesion and axon guidance. In vitro studies have shown that ethanol exposure can disrupt cell motility, and can induce defects in neuronal migration, which includes aberrant neuronal migration within and beyond the cortex (Lindsley et al 2006).

MARS is part of a family of enzymes that play a critical role in protein biosynthesis by charging tRNAs with their cognate amino acids. MARS is a component of the multi-tRNA synthetase complex and catalyses the ligation of methionine to tRNA molecules; it is essential for appropriate translation of mRNA into protein. With regards to the relationship between MARS and PEE, a study using mars (-/-) mutant reported that ethanol interacts with mars to regulate protein synthesis. Untreated mutants were smaller in size than ethanol-exposed or untreated wild type embryos, and ethanol-exposed mutants showed further abnormalities including a reduction in the cartilages from the first 2 pharyngeal arches, as well as in neurocranium size (Swartz et al 2014).

90

In DVD-PEE samples, the top upregulated proteins were La Ribonucleoprotein Domain Family Member 1 (LARP1) and COTL1 (discussed above), while Proteasome 26S Subunit, Non-ATPase 13 (PSMD13) and Proteasome subunit alpha type-5 (PSMA5) were the top downregulated molecules. LARP1 is an RNA-binding protein that promotes translation of specific classes of mRNAs downstream of the mTORC1 complex (Fonseca et al 2015). While there has not been previous reports on the association of vitamin D and

LARP1, there is evidence to show that 1,25(OH) 2D3 is involved in mTOR signalling pathway regulation by stimulating the expression of DNA damage-inducible transcript 4 (DDIT4), which is a potent suppressor of mTOR activity (Lisse et al 2011).The results from the present experiment suggest that vitamin D may regulate other members of the mTOR signalling pathway, whereby LARP1 is a plausible candidate for further investigation. With regards to PEE, a direct association with LARP1 had not been previously reported; however, PEE has been shown to suppress mTORC1 activity in neuronal cells as well as other cell types such as hepatocytes and cardiomyocytes, by inhibiting P70S6K and 4E- BP1, which are used as a hallmark of mTORC1 activation and are correlated with autophagy inhibition (Girault et al 2017).

PSMD13 is a component of the 26S proteasome, a multiprotein complex involved in numerous cellular processes, including cell cycle progression, apoptosis, or DNA damage repair. A lung development study found that PSMD13 was one of 29 proteins dysregulated in lung tissue of vitamin D-deficient mice (Chen et al 2016). Another study in human colon cancer cells reported that 1,25(OH) 2D3 administration can dysregulate the expression of several members of the ubiquitin-proteasome system, including PSMD13 (Álvarez-Díaz et al 2010). In terms of PEE, an association with PSMD13 has not been previously reported; a genetic screen in alcoholic patients versus controls indicated that protein ubiquitination pathway was dysregulated in lymphoblastoid cells from alcoholic subjects, where several molecules, including PSMD13, were aberrantly expressed (McClintick et al 2014). The results from the present study suggest that besides chronic ethanol consumption, PEE may also affect the expression of PSMD13.

PSMA5 is one of the 17 essential subunits that contributes to the complete assembly of 20S proteasome complex. As mentioned above, the proteasome plays a role in several cellular processes that regulate neuronal development. A specific relationship between PSMA5 and vitamin D or PEE has not been previously described; however, pathway

91 analysis from this experiment, as well as reports in the literature, have shown that both DVD deficiency and PEE can affect processes that are also regulated by the proteasome, including cell cycle and apoptosis (Anthony et al 2008, Ko et al 2004).

There were a number of limitations identified in this experiment, which can be addressed in future studies. While the broad approached taken in this assay served to test the biological plausibility of the impact of DVD deficiency and PEE on the regulation of proteins involved in neurodevelopment, top dysregulated candidates require independent validation by either performing another proteomics screen and matching altered molecules to those reported in this experiment, or via Western Blot. Another potential limitation was the tissue selected for this assay, for the reason that cerebrum samples are highly heterogeneous and prevent the identification of altered molecules in specific cell types. Lastly, the assay was performed at a single time-point (P0), which means that it is not known whether the alterations in candidate proteins persist into postnatal stages, or correlate with dysregulated behaviours. This can be addressed by evaluating expression levels of candidate molecules after in postnatal samples collected after behavioural assessments.

4.4 Conclusion

The results obtained in this chapter showed that maternal vitamin D status did not interfere with BACs or ADH levels, suggesting that there was no effect of vitamin D deficiency on the metabolism of ethanol. With regards to gene expression, RNA-Seq analysis first confirmed the effects of DVD and PEE on signalling pathways previously noted in the literature as vulnerable to these exposures. The assay also revealed that the combination group (DVD-PEE) had the most number of affected transcripts at P0, which supports the overarching hypothesis. However, there was no overlap of altered molecules between single hit exposures (DVD or PEE) or their combination, which suggests the effects of DVD-PEE act upon independent pathways from those affected by DVD or PEE alone. Furthermore, a SWATH proteomics screen showed that changes found at transcript level were not translated into protein, because the number of DVD-PEE affected proteins was much smaller and did not coincide with transcripts affected at mRNA level. In agreement with previous literature, these results suggest that DVD deficiency had a strong impact on gene transcription, while PEE had a subtler effect. These effects appeared to be attenuated during protein synthesis stages, resulting in a less severe effect at protein

92 level. Consequently, the effects of DVD deficiency and PEE on epigenetic regulation would be valuable to assess in future studies.

93

Effects of DVD deficiency and PEE on neonatal candidate gene expression and 5 neuroanatomy

94

Chapter 5. Effects of DVD deficiency and PEE on neonatal candidate gene expression and neuroanatomy.

5.1 Introduction

Vitamin D can regulate the expression of genes involved in important neurodevelopmental processes, including cell proliferation, apoptosis, neuronal migration and dopamine signalling (Cui et al 2010, Kesby et al 2009, Ko et al 2004). The absence of vitamin D during crucial stages of development has been shown to disrupt the expression of several transcription factors, some of which have been associated with neurodevelopmental disorders such as schizophrenia and autism. A study using neonatal mouse forebrain reported dysregulation of candidates from the reelin and neuregulin pathways in DVD- deficient samples (Harms 2011). Dysregulated reelin and neuregulin signalling pathways have been associated with neurodevelopmental consequences, including altered brain morphology (neuregulin (O'Tuathaigh et al 2010); reelin (Badea et al 2007)), and behaviours analogous to the positive and negative symptoms of schizophrenia (Ammassari-Teule et al 2009, Krueger et al 2006, Marrone et al 2006, O'Tuathaigh et al 2007, Salinger et al 2003). Another study on BALB/c mice reported foetal neuroanatomical changes and altered neural gene expression as a result of DVD deficiency. Transcripts involved in dopamine regulation, FoxP2 and tyrosine hydroxylase (Th), were reduced in DVD-deficient foetuses, while neuroproliferation markers Bdnf and Tgf-1 were increased, thereby highlighting the crucial role of vitamin D for normal brain development (Hawes et al 2015).

Similarly, PEE has been shown to dysregulate expression of genes important in numerous signalling cascades critical for normal embryogenesis, including the Sonic hedgehod, Wnt and Notch pathways (Ahlgren et al 2002, Li et al 2007, Serrano et al 2010, Tong et al 2013, Vangipuram et al 2012). PEE can also disrupt the regulation of neurotransmitter signalling including glutamatergic, dopaminergic and GABAergic transmission (Naassila et al 2002, Sathyan et al 2007, Valenzuela et al 2011, Zink et al 2009, Zink et al 2011). Focusing on moderate dose of exposure in mice, previous studies have found subtle gene expression changes at embryonic stages (Yamada 2017), further alterations in adolescent offspring (Marjonen et al 2015), and long-lasting changes in adults (Zhang et al 2015). 95

Altered neuroanatomy has been previously reported for DVD-deficient rats and mice. However, results have often been in opposite directions. For example, DVD-deficient rats were found to have increased ventricle size and decreased cortical thickness at birth (Eyles et al 2003). A study in mice found a decrease in lateral ventricle volume in both males and females at embryonic age (E18) using ex-vivo MRI (Harms et al 2012a). Another study using in-vivo MRI on DVD-deficient male mice at two different time points (P210 and P490) showed that DVD-deficient mice had smaller ventricles, while whole brain and hippocampal volumes were unaffected at P210. In control mice, the volume of the hippocampus did not differ significantly between the two ages. However, DVD-deficient mice experienced a significant 20% decline in hippocampal volume from P210 to P490, indicating that age-related neuroanatomical changes may be potentiated in DVD-deficient mice (Fernandes de Abreu et al 2010).

Prenatal ethanol exposure leads to varying levels of cortical dysfunction, including defects in neuronal migration, changes in apoptosis or cell death, alterations in cortical thickness, callosal connectivity, and selective reduction of GABA neurons (Cao et al 2009, Ikonomidou et al 1999, Miller 1986, O’Leary-Moore et al 2011, Sampson et al 1994, Smiley et al 2015). Other studies have identified non-cortical damage to the developing nervous system from prenatal ethanol exposure. For example, Livy and colleagues described abnormal development in the hippocampus, cerebellum, and CC following prenatal ethanol exposure in a rat model of FASD (Livy et al 2008, Livy et al 2003, Maier et al 2001). A recent study reported alterations to developmental cortical thinning across functionally diverse regions of cortex, and altered development of extra neocortical structures, with correlative behavioural deficits in early adulthood, which are consistent with documented human patterns of birth defects present at early developmental stages (Abbott et al 2016).

The main focus of the experiments reported in this chapter was on neonatal subjects, which is an important time point because data collected at this stage occurred prior to vitamin D reintroduction and avoided any potential environmental confounds from altered maternal care, housing or handling. Due to delays with accessing shared facilities, experiments in this chapter were run in parallel to the RNA-Seq and SWATH Proteomics screen discussed in Chapter 4. The main aims of this experiment were: 1) to test the expression levels of candidate genes obtained from previous experiments and the

96

literature; 2) to localize the protein expression of candidate markers in neonatal sections; and 3) to study whether DVD deficiency and PEE have large-scale impacts on the trajectory of brain development an effect on neonatal neuroanatomy, particularly the size and structure of the ventricles and hippocampus.

5.2 Materials and methods

5.2.1 Animals, housing and exposures

Twenty adult male and 40 four-week old female C57BL/6J mice were purchased (ARC, Perth, WA) and housed as described in section 2.2.1. The same sires were used throughout the two cohorts of females to reduce variability from different sires. Dams were randomly allocated to one of four experimental groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) following diet and exposure methods described in section 2.2.2 and 2.2.3.

5.2.2 Neonatal brain tissue collection

Two litters per experimental Group were selected for molecular analyses. Within 12h from birth, pups were decapitated and whole cerebrum tissue was dissected. Left and right hemispheres were collected and snap frozen in liquid nitrogen and then stored at -80˚C.

5.2.3 qRT-PCR

From the brain tissue collected at birth, 44 samples (n=11/Group) were selected for gene expression analysis, as described in detail in section 2.2.5 Briefly, one cerebrum hemisphere from each sample was selected at random, RNA (1µg) was extracted and reverse transcribed. Primers for qRT-PCR analysis of 15 candidate were designed (Appendix 1). RT reactions were diluted 20 times and 3 µl was used in qRT-PCR with 1 x SYBR Green MasterMix and 1 µM of forward/reverse primers. Every sample was loaded in triplicate in each qRT-PCR plate and followed by qRT-PCR analysis. Hprt1 was used as housekeeper. Its expression levels were assessed across all samples using the Ct values from the qPCR reaction and performing a one-way ANOVA to confirm there were no significant changes in expression across experimental groups (Diet: F1,41=0.53, p=0.47;

97

Exposure: F1,41=0.24, p=0.63; DietxExposure: F1,41=0.38, p=0.54). Analysis of was performed using the comparative Ct method (Schmittgen et al 2008).

5.2.4 Immunohistochemistry

a) Neonatal brain perfusions

Within 24h of birth, neonatal mice were anaesthetised by intraperitoneal injection of 0.05ml lethobarb (50:1) and perfused transcardially using a peristaltic pump (MasterFlex, Cole- Parmer, IL). 6ml of 0.9M saline solution was perfused at approximately 5ml per minute, followed by 5ml 4% formaldehyde (FA) in saline solution. Animals were decapitated and skull was not removed in order to visualize craniofacial structure. Brains were immersed for 4 days in 5ml of the same fixative solution used for perfusion. Samples were then transferred into 15mL of saline solution that contained 100µM MnCl2, a contrast agent used in MRI in unmyelinated samples, and were placed at 4°C to incubate.

b) Tissue preparation for paraffin wax embedding

Fixed neonatal brain tissue were post-fixed in 10% piric acid (Sigma-Aldrich, Castle Hill, Australia) in 0.1 M PBS pH 7.2 for 3h. Brain tissues were washed in 0.1 M PBS to remove excess piric acid. Tissues were then processed for paraffix wax embedding according to standard protocols (Bancroft et al 2008). Samples were dehydrated by processing the tissues through a series of increasing concentrations of ethanol, from 50%, 70%, 90% and 100% for approximately 1h each. Tissues were placed into a second solution of 100% ethanol to remove all traces of water. Tissues were then cleared in 100% xylene fir 1h at 25C prior to being placed into warm (60C) paraffin wax (Paraplast, Lomb Scientific, Taren Point, Australia). Tissues were kept at 60C to assist the paraffin wax to penetrate the tissues. Paraffin wax was changed three times for approximately 1h each, to allow tissues to be completely infiltrated with paraffin wax. Tissues were then placed into embedding moulds that contained melted pure paraffin wax to form a block. The paraffin was wax rapidly set at 4C for 5min and wax embedded tissue blocks stored at room temperature.

98

c) Wax-embedded tissue sectioning

Wax-embedded brain samples were sectioned using a Leica rotary microtome RM 2125 (Leica Microsystems, Gladesville, NSW Australia). Sections 10um in thickness were cut and floated onto water in a water bath (40C) to stretch wax embedded tissue sections. Tissue sections were then collected onto silicone-coated glass microscope slides. Sections were dried down onto sides in an oven (35C) and then stored at room temperature.

d) De-waxing and rehydration of wax-embedded tissue sections

Wax sections on glass slides were de-waxed by placing slides into two changes of 100% xylene for 10mi each. Sections were placed into two changes of 100% ethanol for 5min each to remove excess xylene. Sections were placed into 1% H2O2 in methanol for 20min to control endogenous peroxide reactions. Sections were then re-hydrated in 50% ethanol/distilled water (dH2O) for 5min. Sections were completely re-hydrated in 100% dH2O.

e) Antibodies and imaging

Primary antibody was the combination of 4.5 ml of blocking solution, 9 µL of DARPP-32 and 9 µL FOXP2 (1:500 dilution). Excess blocking solution was removed, slides covered with 300 µL of primary antibody solution and left for 16 H in humid environment at room temperature. Slides were then washed three times in PBS for 15 minutes. Species specific secondary antibody solution was the combination of 5 µL of Anti-Rat IgG AlexaFluor(R) 555, 5 µL of Goat AntiRabbit AlexaFluor(R) 488 (Invitrogen) and 5ml blocking solution. Slides were covered in blocking solution followed by 300 µL of secondary antibody and left in a humid environment for five hours at room temperature. Three PBS washes of slides occurred on the shaker for 15 minutes. Slides were placed for one minute in DAPI solution (3 µL DAPI with 15 µL sodium azide). Excess solution was dried; slides covered in Dabco, a cover slip placed on top and sealed with nail polish. Images were taken on Diskovery Spinning Disk, at 60x objective, NIS element selected DAPI, A488 and A568 confocal channels. Images were edited on Fiji 34. FOXP2 and DARPP-32 was compared to DAPI staining to get a qualitative level of expression.

99

5.2.5 Ex-vivo Magnetic Resonance Imaging (MRI)

Fixed neonatal brain samples were superglued by the ventral jaw and immersed in perfluoroether solution (Fomblin Y06/06, Solvey Solexis, Italy). Brains were imaged in the 16.4 Tesla microimaging facility (Bruker BioSpin; Centre for Advanced Imaging, University of Queensland) using a 28mm diameter SAW coil (M2M Imaging, Brisbane) using three- dimensional diffusion-weighted spin-echo images. Diffusion weighting was applied in rostro-caudal direction with b-value = 1000 s/mm^2. The repetition time/echo time (TR/TE) of 400/20ms and diffusion pulse/mixing times of 2.5/12ms with 4 scan averaging. 8 samples were imaged simultaneously with the field of view (FOV) of 32 X 25 x 25 mm at 80µm isotropic resolution. The total imaging time is 16 hours.

3D volumes of 13 brain structures (including , cerebrum, basal ganglia, pineal gland, midbrain-hindbrain, mammillary body, cerebellum, diencephalon, inferior colliculus, superior colliculus, substantia nigra, temporal limb of anterior commissure, fimbria of hippocampus) were measured using registration of P0 MRI atlas (LONI, USC) to the individual brain samples. Briefly, brain masks were created using a semi-automatic segmentation using the program ITK-SNAP. P0 atlas was registered using FSL linear and non-linear image registration (FLIRT-FNIRT) (fmrib.ox.ac.uk). The result of segmentation was carefully inspected and edited using ITK-SNAP prior to obtaining volumetric readouts.

5.2.6 Histology

To assess the morphology of the tissue, selected re-hydrated wax embedded tissue sections were stained using hematoxylin and eosin. Sections were washed in dH2O to remove excess stain and then dried at room temperature. Tissue sections were mounted using glass coverslips and permanent mounting medium DePEX (ProSciTech, Thuringowa, Australia). Sections were examined using a Metafer VSlide Scanner by MetaSystems using Zeiss Axio Imager Z2.

To measure the volume of the areas of interest (ventricles and hippocampus), analysis of images takes as described above was performed using NIH Image J. The regions were manually traced (blind to Group) following reference points to maintain consistency (Fig 23 A,B). Volumes were calculated by converting pixel number to µm (microscope provides ratio), and statistical analysis was performed using SPSS (ver 24) 100

A B

Figure 5.1. Histological analysis of neonatal brain samples. Samples were paraffin-fixed, sectioned (10μm thickness), stained and imaged for neuroanatomical measurements of (A) ventricles and (B) hippocampus.

5.2.7 Statistical analysis

Results were analysed for statistical significance using the SPSS statistics software package (ver. 24, SPSS Inc., Chicago, Illinois). Data were analysed using ANOVA and, where appropriate, repeated measures were used. Student’s t-test was used to assess the difference between two groups. A p value of <0.05 was considered to be statistically significant. In terms of qPCR analysis, there is no established method to test for multiple comparison in experiments where a large number of (potentially unrelated) candidates are assessed. 훼 Bonferroni correction (푝 = ⁄푛 ) is known to be highly conservative, often resulting in increased False Negative findings. To address this, the results from this experiment were analysed with two significance thresholds (uncorrected and corrected).

5.3 Results

5.3.1 Gene expression of selected candidates (qRT-PCR)

The expression of 15 candidate genes (Table 5.1) was assessed in a total of 44 neonatal cerebrum samples (n= 11/Group) via one-way ANOVA. There was a main effect of Exposure in the expression of two candidates. PEE samples (Std or DVD diet) showed a

101

significant downregulation of FoxP2 (F1,40=14.441 p<0.001; Fig. 5.2A). The expression of

Dopamine- and cAMP-regulated phosphoprotein, Mr 32 kDa (Darpp-32) in DVD-H2O samples appeared to be upregulated, however, the effect of Diet was not significant

(F1,40=1.08 p=0.31; Fig. 5.2B). On the other hand, samples exposed to PEE (Std or DVD diet) showed a significant decrease of Darpp-32 expression (F1,40=4.34 p<0.05; Fig. 5.2B). The expression of the remaining 13 candidate genes, including glutamate (Vglut1, Vglut2, Grin3b), dopamine (Th, D2R, Tbr1) and ASD (Cntnap2, Reln, Disc1, Itga3, Itgb1, Itgb3, Mef2c) –related transcripts that were assessed did not show alterations as a result of Diet or Exposure (Fig. 5.3 A-H, Table 5.1). Taking into consideration testing for multiple comparisons, Bonferroni correction was carried out, after which only Foxp2 was dysregulated in PEE samples.

Main Effect of Exposure: (F1,40= 14.441 p<0.001) Main Effect of Exposure: (F1,28= 4.271 p<0.05)

P0 FOXP2 P0 DARPP32 A B

Std-H2O n

n 1.5 1.5

o

o i

i DVD-H O s

s 2

s

s

e

e r

r Std-PEE p

p 1.0 1.0

x

x

e

e

DVD-PEE

e

e

v

v

i

i

t

t a

a 0.5 0.5

l

l

e

e

R R

0.0 0.0

Figure 5.2. Expression levels of affected candidate genes in P0 cerebrum. (A) There was a main effect of Exposure in FoxP2 levels. (B) There was a main effect of Exposure on the expression of Darpp-32. Data presented as mean  SEM, (n=11/Group).

102

A B C D

2.0 P0 TBR1 0.0025 P0 DISC1 2.0 P0 CNTNAP2 2.0 P0 Reelin

n n

n

n

o o

o

o i

i 0.0020

i

i

s s 1.5 s 1.5

s 1.5

s s

s

s

e e

e

e

r r r

0.0015 r

p p

p

p

x

x x

1.0 1.0 x

e e

e 1.0

e

e

e e

0.0010 e

v

v

v

v

i

i

i

i

t

t

t

t

a

a

a

a

l

l l

0.5 0.5 l 0.5

e

e e

0.0005 e

R

R

R R

0.0 0.0000 0.0 0.0

Std-H2O

DVD-H2O Std-PEE DVD-PEE E F G H

2.0 P0 TH 2.0 P0 D2R 2.0 P0 Mef2c 2.0 P0 Vglut1

n

n n

n

o

o o

o

i

i i

i

s

s s

1.5 1.5 1.5 s 1.5

s

s s

s

e

e e

e

r

r r

r

p

p p

p

x

x x

1.0 1.0 1.0 x 1.0

e

e e

e

e

e e

e

v

v v

v

i

i i

i

t

t t

t

a

a a

a

l

l l

0.5 0.5 0.5 l 0.5

e

e e

e

R

R R R

0.0 0.0 0.0 0.0 Figure 5.3. Expression levels of unaffected candidate genes in P0 cerebrum.

103

Table 5.1. List of genes assessed at P0 via qRT-PCR.

p=0.05 p=0.004

DVD- H2O Std-PEE DVD-PEE DVD- H2O Std-PEE DVD-PEE Vglut2 ------Vglut1 ------Grin3b ------Itga3 ------Itgb1 ------Itgb3 ------Th ------

FoxP2 - -

Darpp-32 - - - -

D2R ------Cntnap2 ------Reln ------Tbr1 ------Disc1 ------Mef2c ------

104

5.3.2 Immunohistochemistry

This experiment was performed to visualize the protein expression and the localization of the candidates obtained from qRT-PCR analysis in the neonatal mouse brains of all groups. Due to experimental limitations (lack of viable slices), protein quantification was not able to be performed. The expression of FOXP2 and DARPP-32 was confirmed in neonatal mouse brains from all experimental groups. Based on previous literature, the striatum was chosen as the region of interest to assess FOXP2 and DARPP-32 expression. FOXP2 and DARPP-32 were co-localised in most neurons in the striatum (Fig. 5.4). FOXP2 was found to be highly expressed in the striatum, to a lesser extent within the neuroepithelium. DARPP-32 expression was found in the patch of the striatum, in an inconsistent manner, where some areas would be low or absent from expression and others dense (Fig. 5.4). DARPP-32 expression in the neuroepithelium was at a higher density compared to FOXP2. Some of the limitations in this experiment were that expression levels varied between animals within each Group, as well as within differing densities in sub-regions of the striatum. Additionally, bright staining of DARPP-32 and FOXP2 did not always correlate to high protein expression, but to poor cells staining and high background reading.

Figure 5.4. Localization of FOXP2 and DARPP-32 in DVD-H2O mouse brain at P0. (i) neuroepethelium and (ii) striatum for which neurons which express FOXP2 (yellow arrow) and DARPP-32 (brown arrow), or combined expression (white arrow); pink arrow denotes cells which express neither protein. Tissue was counter stained with DAPI to identify nuclei.

105

5.3.3 Ex-vivo MRI

Brain structure was analysed in 32 neonatal brains from all groups (n=8/Group). Once results were corrected for multiple comparison, using familywise error corrections, there were no significant differences between Diet, Exposure and control groups in the volume of any of the brain regions measured or in the overall volume of the brains (Fig. 5.5) No significant effect of Sex was found, therefore, data were pooled. Ventricle and hippocampal volume were of particular interest in this analysis, however, the neonatal MRI brain atlas used did not have ventricle volume measurement as an option and for automated segmentation and so further histological analysis and manual region of interest measurements was performed to obtain these results.

A P0 MRI 1024 Std-H2O

DVD-H2O Std-PEE 32

) DVD-PEE

2

g

o

L

(

) 3

m 1

m

(

e

m

u

l

o V 0.03125

0.0009765625 lb m ia d in y m n s s ra re s u u l n a d u lo lu lu g u u b r g la r o ll a u u i s p . b n g b .b e h c c n s m y e a . d y li li . i r r g l n r b p l l ia a to e l. a i a re e o o t m c c a e h ll e c c c n m o c s in . i n r. r. a p fa a in c e t o p l p a m i io io s .c i o b r m d r r b r .h b a e e u o f p s ri ia id m in u e r s t b m n m .a fi b m li l. ra o p m e t

Figure 5.5. Neonatal brain anatomy. MRI was performed on neonatal brains from all experimental groups. Thirteen brain regions were measured and no gross volumetric alterations were found as a result of either DVD deficiency, prenatal ethanol exposure or DVD-PEE mice. There was no significant effect of Sex, therefore, data were pooled. Data presented as log 2 mean  SEM, (n=8/Group).

106

5.3.4 Histology

A total of 32 samples (n=8/Group) were processed for histological analysis. Images were manually scored to measure ventricular and hippocampal volume. There was no effect of Group in ventricle size, while there was a main effect of Diet in hippocampal volume

(F1,28=8.316 p<0.05; Fig. 5.6 A,B), whereby DVD deficient animals (H2O or PEE) showed an enlargement in total hippocampal volume.

Main Effect of Diet: (F1,28= 8.316 p<0.05)

P0 Ventricles P0 Hippocampus Std-H2O A B DVD-H2O 80000 600000 Std-PEE

60000 DVD-PEE

)

) 2

2 400000

m

m

m

m

( (

40000

a

a

e

e r

r 200000 A A 20000

0 0

Figure 5.6. Effects of DVD deficiency and PEE on neonatal neuroanatomy. (A) There were no effects of Diet or Exposure in ventricular size of neonatal samples. (B) There was a main effect of Diet, where animals exposed to DVD deficiency (H2O or PEE) showed an increase in hippocampal volume. Data presented as mean  SEM, (n=8/Group).

5.4 Discussion

This chapter focused on neonatal offspring of DVD deficient and PEE dams. Candidates obtained from the literature were assessed for gene expression, and gross neuroanatomy was evaluated. The main findings were 1) candidate transcripts FoxP2 and Darpp-32 were downregulated as a main effect of Exposure (irrespective of Diet); 2) no changes in gross neuroanatomy were found in MRI study; 3) histological analysis revealed an enlargement in hippocampal volume as a main effect of Diet (irrespective of Exposure).

107

5.4.1 Effects of DVD deficiency and PEE on neonatal candidate gene expression

Results from this experiment showed that PEE altered the expression of FoxP2 and Darpp-32 in neonatal mouse brain tissue, suggesting that dopamine signalling is vulnerable to brief, moderate exposure to ethanol. Taking into account testing for multiple comparisons, Bonferroni correction was also carried out, after which only Foxp2 remained as significantly affected. As this was a preliminary assay to test genes of interest, the relevance of both Foxp2 and DARPP-32 in terms of PEE will be discussed below, with the limitation that further validation is required.

Dysregulation of Foxp2 and Darpp-32 is a consistent finding with previous results highlighting the impact of ethanol on the expression of dopamine-related candidates. The precise mechanism by which PEE affects dopamine signalling remains unclear, however, the epigenetic modulation of transcripts, such as those affected in this study, may be a plausible mechanistic explanation that warrant further investigation in future studies. Additionally, the impact of the dysregulation of FoxP2 and Darpp-32 may be addressed by evaluating expression at protein level, and by assessing dopamine-related behaviours, such as communication and sociability.

FoxP2 was initially associated with language, as mutations in this gene resulted in severe speech and language impairment. Disruptions of FoxP2 chromosomal region (7q31) have been linked to a spectrum of neurodevelopmental disorders, such as autism and schizophrenia, as well as ADHD, depression and dyslexia , for the reason that these disorders often present alterations in language endophenotypes (Corominas et al 2014, Gong et al 2003, Li et al 2005, Li et al 2013, McCarthy-Jones et al 2014, Padovani et al 2010, Ribases et al 2012, Sanjuan et al 2006, Schaaf et al 2011, Spaniel et al 2011, Tolosa et al 2010, Wilcke et al 2011), although direct genetic association between FoxP2 and specific disorders (as those mentioned above) have not been found (Gauthier et al 2003, Laroche et al 2008, Newbury et al 2002). Furthermore, FoxP2 expression is not exclusive to humans and is found in several brain regions during development and adulthood. Therefore, rather than FoxP2 being seen as a specific speech gene, it is a complex that is now known to control the expression of numerous targets, suggesting it is involved in multiple processes in brain development and function,

108

including neurite outgrowth and cortical neurogenesis (Chiu et al 2014, Spiteri et al 2007, Tsui et al 2013, Vernes et al 2011, Vernes et al 2007). Moreover, FoxP2 specifically affects distinct stages of neuronal maturation and its expression persists throughout development into adulthood, which suggests it acts in an activity-dependent manner (Ferland et al 2003).

FoxP2 is broadly distributed throughout the mouse brain; it extends to lower cortical layers (majority Layer VI), striatum, mesolimbic and nigrostriatal dopaminergic systems, thalamic somatosensory areas, cerebellum (Purkinje cells), inferior olivary complex, olfactory system and ascending auditory and visual relays (Campbell et al 2009, Ferland et al 2003). This widely distributed expression suggests that FoxP2 interacts with genetic factors that define a number of circuits and pathways, and does not regulate selective circuits involved in verbal or vocal functions (Campbell et al 2009).

During postnatal development, all FoxP2+ neurons in Layer VI of the cortex co-localize with Tbr1, which is a transcription factor, crucial for Layer VI development for the reason that it defines major efferent and afferent innervations. Previous work on Tbr1-/- mutant mice has shown major cortical malformations (Hevner et al 2001). FoxP2 and Tbr1 have also been shown to interact via a T-box domain in Tbr1 (Deriziotis et al 2013, Sakai et al 2011). Furthermore, FoxP2 neurons in layer VI express Tbr1 from P0 to P7. Tbr1 begins to be expressed in layer VI at E11.5, while FoxP2 is initially expressed at E14.5 (Bulfone et al 1995, Ferland et al 2003, Hevner et al 2001). This suggests that Tbr1 may regulate the expression of FoxP2 (at least in layer VI of the developing cortex). Nevertheless, Tbr1 was not altered in DVD deficient or PEE samples, suggesting FoxP2 regulation may be altered through a different pathway, such as via the epigenetic marker MiR-9 (Pappalardo-Carter et al 2013).

FoxP2 has been shown to be upregulated after white noise stimulation in mice, further suggesting its expression is activity dependent (Horng et al 2009). Moreover, studies in FoxP2 mutant mice have shown that dopamine levels are dramatically affected in cortico- striatal structures, while serotonin levels where altered in nucleus accumbens (NAcc) (Enard et al 2009). NAcc is particularly implicated in reward-associated learning, and FoxP2 heterozygous mice show a specific underactivation pattern to Drd1-positive neurons in this region; also, FoxP2-NAcc knockdown mice show attenuation of -

109

induced locomotor behaviour, as well as decreased social preference (Mombereau et al 2013).

Furthermore, work on FoxP2 function during adulthood using songbirds and mice have shown a tight link between FoxP2 and the ability to learn and modify the accuracy of socially driven vocalizations, with the involvement of the dopaminergic system (Murugan et al 2013). For example, adult birds with a FoxP2 knockdown mutation in Area X (homologue to mammalian basal ganglia) show 1) impairments in timing signal sensitivity, which is modulated by dopamine receptor 1 (Drd1), and 2) decreased levels of the key dopamine signalling regulator, DARPP32 (Murugan et al 2013, Teramitsu et al 2006).

Darpp-32 is expressed in the majority of medium-sized spiny neurons (MSN) in the striatum (Ouimet et al 1998, Walaas et al 1984), and is significantly less prominent in other brain regions, such as deep cortical layers. Darpp-32 activity in striatal neurons is associated with modulation of cyclic-AMP (cAMP) (Anderson et al 2005) and has been proven to be fundamental in the modulation of DA-mediated effects in the brain (Walaas et al 1983). Furthermore, Darpp-32 is phosphorylated in response to DA upon dopamine receptor 1 (Drd1) activation, which in turn results in strong inhibition of protein phosphatase-1 (PP-1) and thereby prevents dephosphorylation further.

Darpp-32 is also an important NMDAR regulator. Upon phosphorylation (via cAMP/PKA pathway activation), Darpp-32 inhibits the phosphatase PP1 (Greengard et al 1999). Furthermore, PKA (from cAMP/PKA pathway) phosphorylates NR1, which contributes to enhanced channel activity, while PP1 dephosphorylates this subunit. NR1 activation paired with PP1 inhibition, maintains NR1 subunit in a phosphorylated state. Additionally, it was recently reported that the Darpp-32/PP1 cascade serves as a regulatory mechanism for neostriatal NMDARs in the presence of ethanol, which like other drugs of abuse, increases dopamine release in the neostriatum (Maldve et al 2002).

5.4.2 Effects of DVD deficiency and PEE on neonatal brain anatomy

MRI was performed on neonatal brains from animals of all experimental groups; performing the imaging on neonatal brains allows for the visualization of gross neuroanatomical abnormalities caused by the prenatal exposures excluding any postnatal

110

environmental confounds. Results showed that gross brain anatomy was unaffected by DVD deficiency and PEE, as there was no significant difference in total brain volume and that of 13 broad regions using automated segmentation. Further histological analysis, which allowed for more detailed analysis, revealed no changes in lateral ventricle volumes, and a significant main effect of Diet, showing increased hippocampal volume. These findings were unexpected, based on what has been previously reported in the literature. DVD deficiency in neonatal rats was associated with increased ventricle size and decreased cortical thickness (Eyles et al 2003). Studies in mice have shown a reduction in ventricular size independent of a change in whole brain volume (Fernandes de Abreu et al 2010, Harms et al 2012a), which may suggest that another brain structure has increased in size. DVD-deficient male mice tested at two different time points, P210 and 490 (30 and 70 weeks of age) had smaller ventricles at P210 but not at P490. With regards to the hippocampus, there was a 20% reduction in hippocampal volume between P210 and P490 in DVD-deficient mice, while there was no significant change in controls (Fernandes de Abreu et al 2010), suggesting that DVD deficiency exacerbates age-related neuroanatomical alterations. Harms (Harms et al 2012a) studied both males and females and encountered a decrease in hippocampal volume in neonatal females, which was normalized in adults, potentially indicating that neuronal migration and differentiation at birth may be altered, resulting in an under-developed hippocampus, which may then be corrected by either postnatal development or the reintroduction of vitamin D at birth.

In ethanol exposure studies, an overall ethanol-induced growth retardation was found in GD17 embryos that were exposed to an acute dose of ethanol on GD8. Additionally, disproportionate reductions in the olfactory bulbs, hippocampus and cerebellum were found, as well as an increase in ventricular volumes (Parnell et al 2009). In another study (Parnell et al 2014), a decrease in cerebellar volume and an increase in septal volume was found in animals exposed to ethanol at GD7-10, while a reduction in hippocampal volume and enlarged pituitaries was observed in animals exposed to ethanol at GD12-16. A more recent MRI study on adult mice exposed to moderate PEE (GD0-8) reported a small decrease in total brain volume, and after normalizing for total brain volume, the hippocampus and lateral ventricles of PEE mice were enlarged, while the olfactory bulb was reduced and no changes were found in cerebellar volume (Marjonen et al 2015). These findings highlight an ethanol ‘stage of exposure’-dependent structural brain abnormalities and stress the vulnerability of certain brain regions during the first trimester

111

of pregnancy. However, findings from this experiment were not consistent with previous literature, as no effects of PEE were found on any of the brain regions measured by MRI or histology. One possible explanation for these discrepancies is the age at which scans were taken (neonates vs adults), which suggests the effects of PEE may have a delayed effect in terms of neuroanatomical alterations, whereby changes are undetectable during early postnatal stages and become more noticeable in adulthood. Another explanation could be due to technical limitations found in this experiment, which included low image resolution in P0 MRI scans due to poor contrast caused by low myelination in neonatal brain samples. Further studies are required to optimize the quality of P0 MRI scans and measure specific regions of interest with equivalent accuracy as in adult samples.

5.4.3 Conclusion

The outcomes from this chapter showed that the combination of DVD deficiency and PEE did not result in exacerbated alterations. Gene expression analysis showed that PEE altered the expression of FoxP2 and Darpp-32, transcription factors involved in dopamine regulation, expressed in striatal MSNs. On the other hand, gross neuroanatomy was not affected in PEE samples, while a subtle change in hippocampal volume was found as a result of DVD deficiency. Overall, the results in this chapter suggest that the effects of DVD deficiency and PEE act upon separate pathways in terms of neurodevelopment regulation.

112

Behavioural phenotype of juvenile C57BL/6J mice exposed to DVD deficiency 6 and PEE

113

Chapter 6. Behavioural phenotype of juvenile C57BL/6J mice exposed to DVD deficiency and PEE.

6.1 Introduction

Autism spectrum disorder (ASD) is a neurodevelopmental syndrome, with a prevalence of approximately 1% of the population CDC 2014 (Elsabbagh et al 2012, Kim et al 2011). ASD includes conditions such as autism, childhood disintegrative disorder and Asperger syndrome, and is diagnosed based on behavioural criteria (Happé et al 2008, Landa 2008, Lord et al 2000, Rapin et al 2008). There are no defined biological markers for ASD and the diagnostic manual of the World Health Organization (WHO) requires the presence of core elements in three specific categories for autism diagnosis: abnormal reciprocal social interaction, impaired communication and repetitive behaviours. These elements have been used as guidelines for the development of mouse models of autism, including behavioural tasks designed to maximize relevance to the types of deficits observed in autism patients. such as reciprocal social interaction, social approach, social preference (category 1), ultrasonic vocalizations (USV), urinary pheromones, olfactory habituation (category 2), marble burying, self-grooming and reversal learning (during T maze test) (category 3) (World Health Organization. The ICD‑10 Classification of Mental and Behavioural Disorders).

Developmental vitamin D (DVD) deficiency is a plausible risk factor for ASD (Fernell et al 2010, Grant et al 2013, Grant et al 2009, Pioggia et al 2014, Saad et al 2015). For example, studies in humans have found significantly reduced vitamin D levels in ASD children compared to healthy controls (Pioggia et al 2014, Wang et al 2016b). Vitamin D deficiency has been directly linked with a number of maternal factors that are individually considered risk factors for ASD, including diabetes, preeclampsia, dysregulated steroidogenesis, depression and maternal infection (Aghajafari et al 2013, Merewood et al 2009). Additionally, vitamin D supplementation during pregnancy has been shown to reduce the risk of pregnancy complications (De-Regil et al 2016). Overall, the relationship between vitamin D status and numerous maternal factors suggests that DVD deficiency may modulate or exacerbate the effects of other ASD risk factors. DVD-deficient rats display increased locomotor sensitivity to NMDA antagonists and impairments in cognitive function in adult offspring (Eyles et al 2003, Kesby et al 2006,

114

Overeem et al 2016). However, its effects at earlier time points have not been thoroughly described in relation to neurodevelopmental disorders. One study found altered maternal retrieval behaviour during a maternal isolation test, but showed no changes in ultrasonic vocalizations (Burne et al 2011a). Studies in juvenile DVD-deficient mice focusing on ASD- related domains have not been reported.

Similarly, prenatal exposure to ethanol is considered a risk factor for neurodevelopmental disorders. However, the literature is not clear on the relationship/causality between PEE and autism, though a number of studies have reported some of the effects of PEE on behaviours relevant to neurodevelopment, such as communication, and sociability. A study on PEE rats showed reduced pup vocalizations and increased latency to call (Hashimoto et al 2001, Motomura et al 2002), which may then alter maternal care and have an impact on social behaviour. Previous studies using PEE rats have found impairments in a number of sociability tasks, where PEE males showed altered behaviours in the goal-box task, cagemate interaction and play-fight assessment (Meyer et al 1986).

Dopaminergic dysfunction has been previously implicated in autism (Young et al 1982). DA modulates motor activity, attentional skills, social behaviour, and perception of the outside world, all of which are abnormal in autism. DA-receptor antagonists have shown the most consistent clinical efficacy in controlled trials in autism (Happé et al 1996). Animal models for autism have used the BTBR mouse strain owing to face validity, based on the reliable and robust expression of behavioural phenotypes reminiscent of clinical manifestations of ASD, including social and communication deficits, increased repetitive behaviours and impaired cognitive flexibility (Scattoni et al 2011, Silverman et al 2009, Yang et al 2012). Previous studies have shown substantially blunted striatal and mesolimbic DA neurotransmission in BTBR mice, which was associated with dysfunctional pre- and postsynaptic D2R signalling (Squillace et al 2014). The mesolimbic DA system is comprised of midbrain dopaminergic projections from the ventral tegmental area (VTA) to the NAcc and prefrontal cortex (PFC), structures closely associated with the limbic system (Spanagel et al 1999). The DA dysregulation observed in BTBR mice may play a role in cognitive flexibility deficits observed in this mouse line, which is another behavioural trait that serves as a plausible proxy for restricted interests and repetitive behaviours observed in ASD patients (Squillace et al 2014).

115

The overarching hypothesis for these experiments was that the absence of vitamin D during development would exacerbate the deleterious effects of alcohol exposure, where animals undergoing DVD deficiency and PEE would present a stronger phenotype than those exposed to a single ‘hit’. The first aim of this chapter was to characterize the behavioural phenotype of animals exposed to DVD deficiency and PEE with a focus on autism-related behaviours at early developmental ages starting with basic communication patterns at early developmental stages (P7), followed by locomotion and sociability at a prepubescent age (P21), and later test candidate genes involved in the regulation of these behaviours (Cntnap2, Mef2c). The second aim was to assess the expression of a number of developmentally regulated genes in animals exposed to both exposures. Following from results in neonatal tissue (Chapter 5), FoxP2 and Darpp-32 were of particular interest, along with genes that may modulate these markers, as well as others that play a role in the regulation of DA synthesis (Th, Maoa, Vmat2, Aadc, Drd1, D2R) and glutamatergic signalling (Vglut1, Vglut2).

6.2 Materials and methods

6.2.1 Animals, housing and exposures

Forty adult male and 60 four-week old female C57BL/6J mice were purchased (ARC, Perth, WA) and housed as described in section 2.2.1. The first 20 sires were used throughout the first two waves to reduce variability from different sires and the second group of 20 was used with the last cohort of females. Dams were randomly allocated to one of four experimental groups (Std-H2O; Std-PEE; DVD-H2O; DVD, PEE) following diet and exposure methods described in section 2.2.2 and 2.2.3.

6.2.2 Ultrasonic vocalizations (P7)

A total of 124 pups (8 males, 12 females Std-H2O; 15 males, 12 females Std-PEE; 20 males, 23 females DVD-H2O; 19 males, 15 females DVD, PEE) were tested at P7 in the USVs and maternal potentiation, adapted from (Scattoni et al 2009b). Each pup was placed into an empty cage and assessed for USVs during a 150s recording session (baseline measurement), then reintroduced into the cage with its mother and the littermates for 2.5min. After which the pup was again placed in the clean empty cage for

116 another 2.5min. Latency to first call, total number, type of calls, and difference between second and first trials were recorded.

During the test, an ultrasonic microphone (Noldus Information Technology, Leesburg, VA, USA) sensitive to frequencies between 10 and 125 kHz was suspended 10 cm above the cage. Ultrasonic vocalizations were recorded using Ultravox XT software (Noldus Information Technology, Leesburg, VA, USA). Settings included sampling rate at 250kHz; format bit 16 bit. For acoustical analysis, spectrograms were generated with a time window overlap of 50%. The spectrogram was produced at a frequency resolution of 488Hz and a time resolution of 1ms. A lower cut-off frequency of 35 kHz was used to reduce background noise outside the relevant frequency band to 0dB. Automatic recognition was used to detect total number of calls, which were then –blind to treatment- manually verified and scored for type of call, repertoire from (Scattoni et al 2009a).

A

Figure 6.1. Ultravox XT software.

6.2.3 Social interaction (P21)

At 21 days of age, litters from all four experimental groups (8 males, 14 females Std-H2O;

18 males, 12 females Std-PEE; 27 males, 26 females DVD-H2O; 20 males, 15 females DVD, PEE) underwent a sociability test, adapted from (Crawley 2007). Briefly, testing took place in 2 stages. First, each animal was placed in the centre chamber of the testing arena and locomotor activity was monitored for 5 min. Secondly, a novel animal was placed in one-of-two small, round, wired cages located on each of the side chambers and the pup’s activity was recorded for 5 min. Analysis consisted on evaluating locomotor activity in a

117 novel arena during trial A and time spent near the novel mouse versus the empty wired cage during trial B.

A

Figure 6.2. Social Interaction. (A) Illustration of the 3-chamber social interaction test.

6.2.4 mRNA expression of candidate genes

A total of 32 (n=4/Group/Sex) cerebrum samples collected at P21 were selected for gene expression analysis. Quantitative PCR was performed and analysed as described in section 2.2.5. Hprt1 was used as housekeeper. There were no significant effects of Group on its expression levels (Diet: F1,29=0.20, p=0.66; Exposure: F1,29=0.05, p=0.82;

DietxExposure: F1,29=0.02, p=0.89). The genes that were assessed are listed below (Table 6.1) (primer sequences found in Appendix 1).

118

Table 6.1. List of candidate genes and relevant pathway assessed at P21.

Gene of Interest Relevant Pathway

Vglut1 Glutamatergic signalling

Vglut2 Glutamatergic signalling

FoxP2 Dopamine regulation/ communication behaviours

Darpp-32 Dopamine regulation

Th Dopamine regulation

Maoa Dopamine regulation

Vmat2 Dopamine regulation

Aadc Dopamine regulation

Drd1 Dopamine receptor 1

D2R Dopamine receptor 2

Cntnap2 Autism-related gene

Mef2c Autism-related gene

6.2.5 Statistical analysis

Results were analysed for statistical significance using the SPSS statistics software package (ver. 24, SPSS Inc., Chicago, Illinois). Where no significant Group x Sex interactions were encountered, data were presented pooled for sex. Data were analysed using ANOVA and. Student’s t-test was used to assess the difference between two groups. A p value of <0.05 was considered to be statistically significant.

In terms of qPCR analysis, there is no established method to test for multiple comparison in experiments where a large number of (potentially unrelated) candidates are assessed. Bonferroni correction (p=α⁄n ) is known to be highly conservative, often resulting in increased False Negative findings. To address this, the results from this experiment were analysed with two significance thresholds (uncorrected and corrected).

119

6.3 Results

6.3.1 Ultrasonic vocalizations (P7)

At P7, 124 pups from all groups underwent a maternal separation/potentiation test. Animals from all groups emitted significantly more calls during the second isolation trial than on the first (F1,116=62.002, p=0.000 Fig. 6.3A). There was a main effect of Sex

(F1,116=5.58, p=0.02, Fig. 6.3B) where males made more calls than females. DVD-H2O pups emitted more calls during the second isolation trial compared to controls (Std- H2O), however this did not reach significance (F1,116=3.4, p=0.068, Fig. 6.3C). A one-way ANOVA revealed a significant increase in calls made by Std-PEE male pups compared to controls (Std- H2O) (t=38.5, df=16.77 p=0.05; Fig. 6.3C), while there was no effect in DVD-

PEE pups, and a main effect of Diet x Exposure was also encountered (F1,116=4.3, p=0.04). There was a main effect of Diet x Exposure in five types of calls during the potentiation trial (Fig. 6.4), Downward (F1,116=3.9, p=0.049), Frequency Steps (F1,116=7.6, p=0.007), Single Harmonic (F1,116=11.3, p=0.001), Two-component (F1,116=5.5, p=0.020),

Upward (F1,116=5.9, p=0.017).

120

Main effect of Sex: (F1,116=5.58, p=0.02)

A Maternal Potentiation B USV Males vs Females 100 * 150 Males 80 Trial A * Females

Trial B s

s

l l

l 100

l * a

a 60

C

C

f

f *

o

o

.

. 40 o

o 50

N N 20

0 0 Std-H O DVD-H O Std-PEE DVD-PEE Std-H2O DVD-H2O Std-PEE DVD-PEE 2 2

Males Females C D * Std-H2O 100 100

DVD-H2O

s

s

l

l

l

l a

a Std-PEE

C

C

f

f o

o DVD-PEE

.

. 50 50

o

o

N N

0 0

Figure 6.3. Ultrasonic vocalizations at P7.

Ultrasonic vocalizations were assessed in C57Bl/6J mice at P7 in two 2.5min trials. (A) Rate of calling between trial A and B is significantly different across all groups, showing maternal potentiation. (B) Males from the control and single exposure groups presented a greater number of calls than females during trial B. (C) Exposure to DVD deficiency alone, as well as PEE increases frequency of calling in males; exposure to DVD-PEE had no effect in rate of calling (Data shown is for second trial). (D) Exposure to DVD deficiency, PEE and the combination of both increased the frequency of calling in females, however not significantly.

121

Figure 6.4. Call types quantification at P7.

Ultrasonic vocalizations were classified into eleven different types and then quantified for the potentiation trial in C57Bl/6J mice at P7. (A) Number of calls emitted by males of all experimental groups. (B) Number of calls emitted by females of all experimental groups. Data presented as mean SEM, ^ represents significant main effect of Diet x Exposure p<0.05. n>8/Group/sex.

122

6.3.2 Locomotion (P21)

During the first 5min trial of the sociability task, animals were free to explore the three chambers for 5min and locomotor activity was assessed. PEE males (Std diet) showed significant hyperactivity compared to control males (Std-H2O) (t=5.6, df=76, p<0.001, Fig.

6.5A). Males from the DVD-H2O and DVD-PEE did not show changes in locomotion. Similarly, there was no effect of Diet or Exposure in females.

Males Females A B Std-H2O 1000 * 1000

DVD-H2O

n

n i i 800 800

m Std-PEE

m

5

5

)

) DVD-PEE m

m 600 600

c

c

(

(

e

e c

400 c 400

n

n

a

a

t

t

s

s i

200 i 200

D D

0 0

Figure 6.5. Locomotion at P21. Locomotion was assessed in P21 animals during the first trial of the social interaction task (5min).

(A) Std-PEE males showed hyperlocomotion compared to controls (Std-H2O), whereas males from the DVD-H2O and DVD-PEE groups did not present significant hyperactivity. (B) There were no significant effects of Diet or Exposure in females. Data presented as mean SEM, * p<0.05. n>8/Group/sex.

6.3.3 Social interaction (P21)

At P21, total of 141 pups from all four experimental groups (Waves 2, 4 and 5) underwent the 3-chamber social interaction test. There was a main effect of Sex x Exposure, where male pups prenatally exposed to ethanol (Std or DVD diet) failed to show a preference for the stimulus mouse vs the empty cup (trial B), whereas control (Std- H2O) animals spent significantly longer time interacting with the stimulus mouse (F3,137=8.279, p=0.004; Fig. 6.6 A) than in the empty side. There was no effect of Diet or Exposure in females (Fig. 6.6 B).

123

A Males B Females

p

p u

u *

c

c

200 Empty 200

y

y t

t * p p Novel * *

* m

m * E

E 150 150

s

s

v

v

e

e s

s 100 100

u

u

o

o

m

m

l

l e

e 50 50

v

v

o

o

N

N

e

e 0 0

m

m i

i Std-H2O DVD-H2O Std-PEE DVD-PEE Std-H2O DVD-H2O Std-PEE DVD-PEE T T

Figure 6.6. Social Interaction at P21.

Sociability was assessed in C57Bl/6J mice at P21 in three 5min trials (habituation, social interaction, social novelty). (A) Std- H2O males spent significantly longer time with the novel mouse than in the empty side, while Std-PEE and DVD-PEE pups failed to show a preference towards the novel mouse. (B) Females from all groups showed preference towards the novel mouse compared to the empty side. Data presented as mean SEM, * p<0.05. n>8/Group/sex.

6.3.4 mRNA expression of candidate genes

The expression of twelve candidate genes was assessed in cerebrum tissue samples collected at P22. Following from results obtained at P0, FoxP2 was of particular interest, however, its expression was not altered at P21. The other gene of interest, Darpp-32, showed a main effect of Exposure, whereby PEE (Std or DVD diet) animals showed increased levels of Darpp-32 (F1,28= 7.859 p<0.05, Fig. 6.7 A,D). Additionally, D2R was increased in DVD animals (H2O or PEE) (F1,28= 8.074 p<0.05, Fig. 6.7 B,E). There was no significant effect of Group on TH expression, however, there was a main effect of Sex on the expression of this gene (F1,28= 7.971 p<0.05, Fig. 6.7 C,F). The expression of the remaining 8 candidate genes, including glutamate (Vglut1, Vglut2), dopamine (Maoa, Vmat2, Aadc, Drd1) and ASD (Cntnap2, Mef2c) –related transcripts that were assessed did not show alterations as a result of Diet or Exposure (Table 6.2). Considering testing for multiple comparisons, a more conservative analysis method was carried out, following the same fashion as in Chapter 5. None of the candidates that were significantly altered via one-way ANOVA were significantly different using a corrected p value.

124

Main Effect of Exposure: (F1,28= 7.85 p<0.05) Main Effect of Diet: (F1,28= 8.07 p<0.05) Main Effect of Sex: (F1,28= 7.97 p<0.05)

P21 DARPP32-Males P21 D2R-Males P21 TH-Males A B C 4 4 4

Std-H2O

n

n n

o

o o

i i

i DVD-H O

s s

s 3 3 3 2

s

s s

e

e e

r r

r Std-PEE

p

p p

x x

x 2 2 2

e

e e

DVD-PEE

e

e e

v

v v

i

i i

t

t t

a

a a

l l

l 1 1 1

e

e e

R

R R

0 0 0

P21 DARPP32-Females P21 D2R-Females P21 TH-Females D E F

4 4 4

n

n

n

o

o

o

i

i

i

s s

3 3 s 3

s

s

s

e

e e

r

r

r

p

p

p

x x

x 2 2 2

e e

e

e

e

e

v

v

v

i

i

i

t

t

t

a

a

a

l l

l 1 1 1

e

e

e

R

R R

0 0 0

125

Figure 6.7. Expression levels of affected candidate genes in P21 cerebrum. (A, D) There was a main effect of Exposure in Darpp-32 levels. (B, E) There was a main effect of Diet on the expression of D2R. (C, F) There was a main effect of Sex on the expression of TH. (D-L) (n=4/Sex/Group). Data presented as mean SEM. n=4/Group/sex.

126

Table 6.2. Expression levels of unaffected candidate genes on P21 cerebrum.

Std-H2O DVD-H2O Std-PEE DVD-PEE

Males Females Males Females Males Females Males Females (n=4) (n=4) (n=4) (n=4) (n=4) (n=4) (n=4) (n=4) Transcript Mean SEM Mean SEM Mean SEM Mean SEM Mean SEM Mean SEM Mean SEM Mean SEM Vglut1 1.000 0.375 1.000 0.495 1.158 0.258 0.765 0.254 0.740 0.239 0.661 0.237 0.905 0.235 1.651 0.284 Vglut2 1.000 0.527 1.000 0.292 1.127 0.517 1.283 0.188 1.449 0.493 1.245 0.230 0.859 0.395 3.431 1.164 FoxP2 1.180 0.310 0.820 0.310 1.590 0.320 1.280 0.330 1.380 0.210 1.230 0.200 1.650 0.200 2.040 0.570 Maoa 1.000 0.106 1.000 0.288 1.148 0.157 1.513 0.168 1.465 0.300 1.281 0.204 1.110 0.127 1.215 0.200 Vmat2 1.000 0.430 1.000 0.293 1.430 0.231 1.100 0.173 1.072 0.338 1.082 0.197 1.404 0.316 1.523 0.300 Aadc 1.000 0.566 1.000 0.460 1.302 0.260 1.247 0.136 1.210 0.332 1.451 0.496 1.446 0.323 2.101 0.537 Drd1 1.000 0.273 1.000 0.299 0.918 0.142 1.204 0.112 0.903 0.072 1.018 0.182 0.980 0.147 1.340 0.449 Cntnap2 1.080 0.150 0.920 0.390 0.960 0.460 0.970 0.100 1.140 0.220 1.070 0.090 1.240 0.180 1.550 0.310 Mef2c 1.000 0.305 1.000 0.396 1.675 0.228 0.877 0.084 1.035 0.264 0.574 0.059 1.752 0.510 1.293 0.192

.

127

6.4 Discussion

The main behavioural findings (P7-21) were increased calling rate in PEE male pups, altered distribution of call types amongst both DVD and PEE groups, and increased locomotion and impaired social interaction in prepubescent PEE males. No behavioural alterations were found in female mice when compared to the control group. Furthermore, expression of two genes was altered, Darpp-32 was increased as a main effect of Exposure, while D2R was increased by DVD deficiency, neither of them showed an effect of Sex. While there was no significant effect of Group, there was a main effect of Sex on TH expression. These results indicate that DVD deficiency did not increase the offspring vulnerability to PEE, but rather partially reversed the phenotype observed in mice with a single exposure to PEE.

Table 6.3. Summary of Behavioural Phenotype in Juvenile Offspring.

Test DVD- H2O Std-PEE DVD-PEE ♂ ♀ ♂ ♀ ♂ ♀

Ultrasonic - - - - - Vocalizations Locomotion - - - - -

Social Interaction - - - -

Darpp-32 - - - -

D2R - -

6.4.1 DVD deficiency and PEE on ultrasonic vocalizations

Previous DVD deficiency studies on USVs in mice are lacking. One study in rats examined neonatal isolation-induced pup ultrasonic vocalizations and maternal–infant interactions using a pup-retrieval test in DVD deficient and control rats. DVD deficient dams retrieved

128 their pups sooner than control dams and engaged in more pup directed activities (sniffing and carrying pups). However, no significant effect of maternal diet was found on the calling rate of isolation-induced USVs by pups (Burne et al 2011b). These findings are in agreement with those found in the present study, where DVD deficient pups did not show alterations in the rate of calling compared to controls (Std-H2O); additionally, there was no effect of DVD deficiency combined with PEE in the vocalization rate.

With regards to ethanol exposure, mixed results have been reported. Marino et al (2002) conducted a study on perinatal ethanol exposure on USVs and maternal-infant interactions and found that maternal behaviours at P2, 4, 6, 8 and 10 were unaffected by ethanol exposure to the dam or the pups. However, vocalizations examined at P5 revealed a significant increase in total calls by ethanol-exposed pups compared to controls, regardless of the isolation condition. On the other hand, others have reported a decrease in the rate of calling at various time points (P3,9,14). For example, a study looking at the effects of ethanol and cocaine exposure in rat pups showed that cocaine had no effect on USVs, while ethanol or ethanol and cocaine caused a deficit in the rate of calling across a 6min isolation test (Barron et al 2005). The altered vocalization response of ethanol- exposed pups might be an underlying factor in the persistent effects of prenatal ethanol exposure on social behaviour. Further, sonographic analysis of nine different call types revealed a selective effect of ethanol on certain waveforms rather than an overall reduction across all types (Barron et al 2005). Detailed sonographic analysis provides a deeper insight while studying USVs, as others (prenatal malnutrition) have reported alterations in waveform topography but not on the frequency of vocalizations (Tonkiss et al 2003).

Neonatal mice make both simple and more complex types of USVs, but their specific function is still unclear. However, it is possible that more complex calls are of particular importance in maternal-offspring interactions, as sounds are easier to locate if the call consists of multiple sound frequencies. This supports preliminary findings in our study, where multi-frequency calls, including Two Component, Single Harmonic and Frequency Steps, were particularly altered in Std-PEE and DVD-PEE pups. Prior research on PEE has shown a delay in PEE pup retrieval by their own dam or a control dam, which suggests that PEE pups may not effectively communicate their needs to their dam, causing a deficit in maternal and pup-directed behaviours that other pups do by vocalizing. D’Amato’s (2001) study suggests that the amount of USV’s emitted by the pup is modulated by the dam’s promptness to respond to the pups needs, therefore, if pups do

129 not respond appropriately either during isolation or in the presence of the dam, it would have a further impact on maternal behaviour, which in turn can have long-term adverse effects (Fleming et al 1999, Ogilvie et al 1997, Ward et al 2002) on adult sexual behaviour, hypothalamic-pituitary adrenal axis development (Liu et al 1997, Moore et al 1997), as well as learning and memory (Barbazanges et al 1996, Lévy et al 2003).

6.4.2 DVD deficiency and PEE on social interaction

DVD deficiency studies on social behaviour are very limited, and no alterations have been reported in rats (Burne et al 2004b). Both clinical and animal studies have shown that ethanol exposure may have detrimental effects on social behaviour. There are multiple ways to assess social domain deficits in rodents. For example, a study on the effects of developmental exposure to ethanol assessed social behaviour in rats at P30 using the alley maze, containing a cage mate in a “goal box”. Animals underwent various forms of isolation and the latency to reach the goal box, as well as the social behaviour once inside the goal box were examined. The study showed that PEE rats ran faster than control rats towards the goal box, and exhibited aberrant social interactions with their cage mate, including increased anogenital sniffing, chasing, hopping and darting, which after accounting for motivational state and social learning, might indicate increased responsiveness to social stimuli (Lugo et al 2003). Another study examined play-fighting behaviour in juvenile PEE rats and found a reversed pattern to the usual sexually dimorphic behaviour. Males normally engage in play behaviour more than females, however, PEE males play-fought less than controls, while PEE females played more than female controls (Meyer et al 1986). Similarly, a study conducted in artificially reared rats (postnatal ethanol administration), found a decrease in active social interactions in males and a decrease in females (P90-95) relative to sex-matched controls, which is the opposite of this sexually dimorphic behavioural pattern (Kelly et al 1994). Other behaviours were also altered in the same sexually dimorphic pattern, including saccharin preference, performance in the Lashley III maze (McGivern et al 1988), which could be mediated by sex differences in the effect of ethanol exposure, with some male systems being more sensitive to ethanol exposure, different to those vulnerable systems in females. Results from our experiments are somewhat in agreement with these findings, given that PEE male pups presented a social deficit in the 3-chamber social interaction test, while females were unaffected.

130

6.4.3 DVD deficiency and PEE on gene expression

Gene expression analysis on a list of candidates relevant to ASD, glutamatergic and dopaminergic signalling revealed that DVD deficiency did not exacerbate the effects of PEE in the expression of the selected genes. There was a main effect of DVD deficiency on the expression of D2R, and a main effect of PEE on Darpp-32 expression, though in the opposite direction to that observed at birth. While none of these candidates passed the Bonferroni correction, they are plausible candidates that may indeed be vulnerable to the effects of DVD deficiency and PEE when assessed in a tissue specific manner and across various time points. Therefore, their biological significance with respect to DVD deficiency and PEE will be discussed below.

DA is a neurotransmitter from the catecholamine family, along with norepinephrine (NE) and epinephrine (EPI). DA modulates processes and behaviours that are altered in individuals with ASD, including cognitive processes (Hughes et al 1994, Mostofsky et al 2000), motor functions (Vilensky et al 1981) and emotional regulation (Gaigg et al 2007). Moreover, the striatum is the major input structure of the basal ganglia (composed of nuclei such as striatum, globus pallidus, and subthalamic nucleus); the basal ganglia process cortical information via either the direct or indirect pathway. Nigrostriatal DAergic neurons modulate striatal activity by different DA receptors expressed by GABAergic medium spiny neurons (MSNs) from these pathways. DA receptors are defined by their effect on adenylate cyclase activity (Kebabian et al 1979)- D1 class encompasses D1 and D5 receptors, these are coupled to stimulatory G-proteins that increase adenylate cyclase activity, while D2 class of receptors includes D2, D3 and D4 receptors, which are coupled to G-proteins that decrease adenylate cyclase activity (Svenningsson et al 2004). Furthermore, the opposing downstream effects of D1 and D2 receptor activation are mediated by Darpp-32, which is a critical modulator of DA signal transmission and controls a range of downstream physiological effects (Svenningsson et al 2004). DA D1 and D2 receptors, as well as Darpp-32 are highly expressed in MSNs in striatum (in mice, rats, non-human primate and human brain) (Bergson et al 1995, Levey et al 1993, Ouimet et al 1992), whereby D1 receptors are found in striatonigral neurons of the direct pathway, D2 receptors are found in striatopallidal neurons of the indirect pathway (Gerfen et al 1990), and Darpp-32 is found throughout the striatum (Ouimet et al 1992).

131

Moreover, studies in rats have consistently shown that DVD deficiency alters dopamine signalling. For example, adult female DVD rats are more sensitive to the locomotor enhancing effects of amphetamine, a dopamine-releasing agent (Kesby et al 2009). Adult DVD-deficient rats were also more sensitive to the dopamine receptor antagonist haloperidol (Kesby et al 2006). In neonatal forebrain tissue, DVD deficiency has been shown to reduce levels of expression of the dopamine-metabolizing enzyme catechol-O- (COMT). Additionally, DVD deficiency was shown to alter the ratio of major dopamine metabolites dihydroxyphenylacetic acid/homovanillic acid (DOPAC/HVA), suggesting there is abnormal dopamine turnover in the neonatal forebrain (Kesby et al 2009). HVA concentrations in urine and plasma have been reported abnormal in ASD patients (Kaluzna-Czaplinska et al 2010, Toda et al 2006). Furthermore, Nurr 1 and p57Kip2- factors involved in early post-mitotic specification of dopaminergic neurons, were reduced as a result of DVD deficiency in the mesencephalon (Cui et al 2010). Overall, there is strong evidence to suggest vitamin D plays an important role in the maturation of dopaminergic systems. Furthermore, a two-hit model study combining hypovitaminosis D and high fat diet found that low vitamin D in conjunction with high fat diet resulted in a blunted locomotor response upon acute amphetamine administration, as well as an increased consumption of liquid amphetamine compared to non-deficient controls. Additionally, calcitriol treatment led to transcriptional changes of DA-related genes in the midbrain and their target neurons in the striatum (which express VDR protein) of naïve mice, enhanced amphetamine-induced DA release and increased locomotor activity after acute amphetamine treatment, thereby providing further evidence that vitamin D3 signalling is capable of modulating DA circuits (Trinko 2013).

With regards to PEE, previous literature has shown alterations in neurobehavioural functions such as attention shifting, movement and spatial working memory (Herrero et al., 2002), which are regulated by striatal circuits. Furthermore, studies in rhesus monkeys have shown disrupted DA-dependent functions including orientation, motor maturity and state control neonatal scores in monkeys prenatally exposed to moderate alcohol during early gestation (Schneider et al 2005). Additionally, moderate PEE resulted in long-term increases in prefrontal dopamine Drd1 binding in male monkeys (Converse et al 2014), and altered D2R levels in a time-dependent manner, where PEE animals exposed during early gestation showed a reduction in DA D2R binding, while those exposed during mid to late gestation showed the opposite effect, reiterating the significance of timing of exposure in alcohol’s long-term effects (Schneider et al 2005). In rats, studies have shown that PEE

132 results in a reduction in dopaminergic activity in the ventral tegmental area (VTA), correlated with hyperactivity and impulsivity (Muñoz-Villegas et al 2017). Another study looking at the effects of PEE on the interaction of the HPA and dopamine systems, which are known to have overlapping neurocircuitries, found that PEE alters both HPA and DA activity and regulation, resulting in increased HPA tone and an overall reduction in tonic DA activity in a sexually dimorphic pattern (Uban et al 2013).

With regards to effects of Sex, tyrosine hydroxylase (TH), which is the rate-limiting enzyme for dopamine synthesis, was overexpressed in female mice. Previous studies in drosophila have shown that TH levels were altered in response to stress, dependent on sex, age and reproductive status of the population (Neckameyer et al 2005), and dopaminergic signalling in response to stress analysis showed that pre- and postsynaptic neurons are recruited into the dopaminergic stress response circuitry in a sexually dimorphic pattern (Argue et al 2013). DVD studies in rodents have shown a sexually dimorphic effect in the number and location of dopamine neurons, and males have lower levels of TH compared to females (Hawes et al 2015), which in turn is regulated by oestrogen in the mouse midbrain. Male and female brains appear to have a different susceptibility to DVD deficiency in early development, and these findings warrant further scrutiny in future studies.

6.4.4 Conclusion

In conclusion, the results from this study suggest that vitamin D deficiency during pregnancy did not exacerbate the effects of short-term, low-dose ethanol exposure. Further, PEE alone induced more prominent effects on behaviour and gene expression, where males appear to be more vulnerable than females, showing disrupted communication and sociability, as well as dysregulated expression of the DA-related gene, Darpp-32. In terms of the DVD-PEE group, it is possible that the transient depletion of vitamin D throughout gestation altered the regulation of signalling pathways involved in the behaviours assessed in this experiment, however, an overcompensation may have been triggered by the reintroduction of vitamin D at birth, resulting in a more “normalized” behavioural phenotype.

133

7 General Discussion

134

Chapter 7. General Discussion.

7.1 Introduction

In this thesis, I have shown that the effects of DVD deficiency in conjunction with PEE have a temporal pattern, whereby gene expression at birth was further dysregulated in DVD-PEE pups compared to DVD or PEE alone. This is shown graphically as a speculative trajectory to summarise the main findings of each of the 4 groups used in this thesis (Fig. 7.1). However, these alterations appeared to be transient because analysis did not correlate with mRNA findings (Chapter 4). Additionally, the regulation of specific candidates of interest was not dysregulated in the combination group (Chapter 5). Furthermore, the synergistic effects of these exposures seemed to dissipate with increasing offspring age.

Figure 7.1 Speculative trajectory of the effects of DVD deficiency and PEE in the offspring. Schematic interpretation of the temporal effects of DVD deficiency (blue), PEE (orange) and DVD- PEE (green) compared to controls, Std-H2O (grey) across the offspring’s lifespan. DVD deficiency resulted in the dysregulation of several genes and subtle neuroanatomical changes at birth, upregulation of DA receptor expression at P21, and no behavioural alterations during prepubescence or adulthood. PEE induced subtle changes in gene expression and did not affect gross neuroanatomy at birth. Male PEE mice showed altered juvenile behaviour; however, these effects dissipated with age, because more subtle changes were identified during adulthood. The two-hit group (DVD-PEE) showed significant changes in gene expression at birth, however, alterations were not persistent in later postnatal stages, given that there were no alterations in juvenile or adult behaviours and gene expression.

135

The overarching hypothesis of this thesis, stating that combining DVD deficiency and PEE would exacerbate the effects of a single exposure in the offspring was initially supported by gene expression changes identified in a broad RNA sequencing screen in neonates (Chapter 4). However, these effects were not persistent in later postnatal time points, during which no additive effects of DVD-PEE were found in juvenile or adult behaviour and candidate gene expression. DVD deficiency altered gene expression (Chapter 4) and hippocampal volume of neonatal mice; in prepubescent DVD-deficient mice, there was a subtle alteration in the expression of a DA-related candidate, yet juvenile (Chapter 5) and adult behaviours (Chapter 6) were not affected. Male PEE subjects were found to be the most vulnerable group, showing alterations in gene expression and behaviour at several time points, including P0, P7, P21 and P70. Transcription factors FoxP2 and Darpp-32 were dysregulated in PEE samples at P0 (Chapter 4); Darpp-32 expression continued to be altered in juvenile samples (P21) of PEE male offspring (Chapter 5) and Vglut2 expression was dysregulated in hippocampal samples of adult (P80) PEE offspring (Chapter 2). In terms of behaviour, male PEE pups showed disruptions in communication (P7), locomotion and sociability (P21) (Chapter 5); locomotion continued to be altered in adulthood (P70), however, learning and memory, as well as MK-801-induced hyperlocomotion were not affected (Chapter 6). These data suggest that the combination of DVD deficiency and PEE transiently increases the offspring’s vulnerability, while DVD deficiency alone produces a subtle phenotype and PEE has a longer-lasting impact at a molecular and behavioural level.

7.2 Impact of DVD deficiency and PEE on neonatal outcomes

The duration of vitamin D deficiency has been explored in previous studies, intending to identify a ‘critical window’ during which the absence of vitamin D exerts its most potent effects. In rats, offspring from dams that were vitamin D deplete until birth had increased lateral ventricle volume at birth (Eyles et al 2003), while if the vitamin D deficiency period was prolonged until weaning, the ventricle enlargement phenotype persisted into adulthood (Féron et al 2005). However, prolonged vitamin D deficiency resulted in hypocalcaemia, which confounded the interpretation of postnatal findings. Therefore, in this thesis, we used a ‘classic’ DVD deficiency model, in which maternal vitamin D deficiency was maintained from conception, through gestation and culminated at birth. Experiments examining gene and protein expression, and brain structure were assessed in neonates, which allowed for the assessment of the effects of DVD deficiency prior to the

136 reinstatement of vitamin D in the offspring’s diet, and prevented potential confounds, such as compensatory effects driven by the reintroduction of vitamin D.

With regards to PEE, timing of exposure has also been shown to have a significant impact on the results observed at gene, protein and behavioural levels. The exposure window utilised in this thesis is developmentally equivalent to the first 3 – 4 weeks of a human pregnancy (Otis et al 1954), a period of time during which most Australian women have yet to confirm their pregnancies (Callinan et al 2012). Central Nervous System (CNS) development is characterized by vulnerable periods during which exposure to teratogens may result in abnormalities specific to the ontogenic events occurring at the time of exposure (Rice et al 2000). Ethanol appears to interfere with all neurodevelopmental stages (Guerri 1998). The first trimester in humans (GD1-11 in mice) is considered the first critical period of development, during which organogenesis begins and the neural tube and crest are formed (Rice et al 2000). Moreover, during the second week of gestation in mice (first month in humans), specific areas of the CNS begin to form with neurogenesis and migration of cells in the forebrain, midbrain, and hindbrain (Rice et al 2000). Mice exposed to acute doses of ethanol during GD 7 or 8 exhibit the craniofacial anomalies associated with FAS, forebrain deficiencies including hypoplasia or aplasia of the corpus callosum, and deficiency in the hippocampus and the anterior cingulated cortex (Marino et al 2004). Previous studies using moderate levels of exposure (the same as that used in this thesis) have shown more subtle changes in behaviour (Sanchez Vega et al 2013), brain structure (Marjonen et al 2015), craniofacial morphology (Kaminen-Ahola et al 2010a) and transcription and epigenetic state in various postnatal tissues (Kaminen-Ahola et al 2010a, Marjonen et al 2015, Zhang et al 2015) Another study evaluated the effects of PEE shortly after the termination of ethanol exposure (GD9) to assess whether the alterations observed at postnatal stages were present from the time of exposure, or if there was a latency in their onset. Findings at GD9 did not reveal changes of a large effect size at this stage (Yamada 2017), which suggests that there is a delayed onset of PEE-induced alterations.

While there are some prior data on DVD deficiency in neonates, the effects of moderate PEE, or the combination of exposures, have not been described. An unbiased screen of mRNA expression in neonatal brain samples showed that DVD deficiency dysregulated transcripts involved in several neurodevelopmental processes, primarily related to molecular transport, cell death and survival, which is consistent with previous reports from

137 in-vitro and in-vivo studies (Eyles et al 2009, Ko et al 2004). Meanwhile, few transcriptional changes were found as a result of PEE, which is consistent with previous assays during embryogenesis (Yamada 2017) and further supports the hypothesis that effects of PEE have a delayed onset. Moreover, the current study characterized the gene expression pattern of DVD-PEE samples for the first time. Changes in gene expression were found to be exacerbated by the combination of exposures, whereby transcripts regulating neurodevelopmental processes such ash cell function and maintenance, cell assembly and organization, and cell signalling were dysregulated.

A novel finding was that circadian rhythm signalling was the top canonical pathway dysregulated by DVD-PEE, whereby specific members of this pathway were significantly downregulated, including NR1D1, NR1D2 and PER3. There is limited evidence for associations between DVD deficiency or PEE and these markers, however, in-vitro studies of calcitriol supplementation have reported that 1α,25-(OH)2D3 administration results in the upregulation of two genes that are part of the circadian rhythm pathway (BMAL1 and PER2). This suggests that vitamin D may have an important role in the maintenance of circadian rhythms at the cellular level (Gutierrez-Monreal et al 2014). Moreover, based on the well-established evidence that exposure to ethanol during crucial developmental periods may result in cell loss, altered pathway coordination between brain regions and disrupted neurotransmitter synthesis, it is reasonable to speculate that ethanol may alter the development of the suprachiasmatic nuclei (SCN), known as the ‘internal biological clock’, which is responsible for the generation of all circadian rhythms and their synchronization (Earnest et al 2001). Although it is unlikely that the damage to the SCN was severe enough to completely disrupt circadian rhythmicity, ethanol could potentially induce more subtle disturbances that would be important to evaluate in future studies, such as light/dark cycle activity assessment.

In addition to evaluating gene expression levels, an unbiased proteomics screen was performed in neonatal samples. It was predicted that protein expression of candidates dysregulated at mRNA level with the largest fold change would also be altered. However, the proteins with the most significant difference in expression did not match the candidates found to be disrupted at mRNA level. Also, given that the number of disrupted proteins was equivalent across all comparison groups (DVD, PEE, DVD-PEE) and the fold change of altered proteins was actually smaller in the combination group than in each of the single-hit comparisons, the exacerbated effect of DVD-PEE observed in gene expression

138 did not persist to protein level. Tight junction signalling, responsible for internal cell signal transmission to modulate gene expression and cell behaviour (Matter et al 2003), was the main canonical pathway found to be disrupted in DVD-PEE samples. While there is no previous data on DVD deficiency and tight junction signalling in terms of neural development, cancer studies have suggested that vitamin D signalling (via VDR) may regulate the level and localization of tight junction proteins in the intestinal epithelium, leading to a more secure intestinal barrier (Fleet et al 2012). Additionally, 1α,25(OH)2D3

(via VDR) has been shown to upregulate the expression of tight junction proteins claudins- 2 and -12 in enterocytes in vitro and in vivo. Since claudins are suggested to facilitate paracellular Ca2+ transport, the role of vitamin D on their expression highlights a novel mechanism behind vitamin D-dependent Ca2+ homeostasis (Fujita et al 2008b). With regards to PEE, a previous study looked at two mouse substrains (C57BL/6J /6N), and performed a broad screen of PEE embryos subjected to acute alcohol exposure (GD8). They found that tight junction, focal adhesion, and adherens junction pathways were the most significantly affected in both substrains, while regulation of the actin cytoskeleton, Wnt signalling, and programmed cell death appeared at lower confidence (Green et al 2007). These findings are consistent with the hypothesis that ethanol's developmental effects may be mediated by common signalling pathways linking receptor activation to cytoskeletal reorganization (Lindsley et al 2006).

Another aspect of development that was evaluated at birth was neuroanatomy. Ex-vivo MRI did not reveal any gross abnormalities in brain morphology of DVD, PEE or DVD-PEE subjects. This was not consistent with prior studies, where DVD deficiency has been shown to alter ventricular volume (Harms et al 2012a), and PEE to induce craniofacial abnormalities (Kaminen-Ahola et al 2010a), as well as hippocampal, ventricular and cerebellar volume alterations (Marjonen et al 2015), although not at birth. Upon further inspection, histological analyses revealed a subtle enlargement in hippocampal volume of DVD samples (total brain volume remained unchanged), which had not been previously encountered in neonates but suggested that learning and memory may be affected in DVD-deficient offspring, as it had been previously described in adult DVD-deficient mice (Fernandes de Abreu et al 2010) (adult behaviour discussed below). There is no preliminary evidence on the morphological effects of DVD-PEE, however, it was hypothesized that subtle changes such as those previously observed in DVD mice, would be exacerbated by the teratogenic effects of ethanol. Nevertheless, animals subjected to both exposures did not show neuroanatomical alterations, which suggests

139 that the effects of DVD deficiency and PEE are not synergistic. One possible explanation for these findings could lie on the opposing effects of vitamin D and ethanol on apoptosis. Cancer studies have shown that vitamin D has a pro-apoptotic role, consequently suggesting that the absence of vitamin D results in upregulated cell proliferation (Fleet 2008). On the other hand, ethanol exposure is known to exert teratogenic effects, which includes increased cell loss. For example, a study found that PEE results in elevated expression of activated Caspases 8 and 3, and that hippocampal mossy cells are particularly sensitive to ethanol-induced neuroapoptosis (Wang et al 2015).

7.3 Impact of DVD deficiency and PEE on early postnatal outcomes

The effects of DVD deficiency and PEE were assessed in terms of behaviours known to be vulnerable in neurodevelopmental disorders, including communication, locomotion and sociability. It was hypothesized that juvenile DVD-PEE mice would display a more severe phenotype in comparison to mice individually exposed to DVD deficiency or PEE. However, DVD-PEE mice showed no alterations in any of the behavioural measures that were evaluated. The total number of USVs during a maternal potentiation test was used to assess communication in young pups (P7). DVD deficiency did not have a significant impact on this behaviour. This is consistent with a previous study in DVD-deficient rats, which reported no changes in USVs in neonates (Burne et al 2011a). On the other hand, PEE male pups emitted a significantly larger number of calls compared to controls, which is in agreement with previous rat PEE studies (Marino et al 2002). The precise circuitry and neurochemistry that mediates this behaviour is not yet known, although several and neurosteroids have been shown to modulate rodent USVs, including dopamine, glutamate, GABA, serotonin norepinephrine and corticosteroids (Branchi et al 2001, Carden et al 1991, Farrell et al 2002, Insel et al 1989, Joyce et al 1999, Kehoe et al 2001, Nazarian et al 1999, Ricceri et al 2007, Winslow et al 1990, Zimmerberg et al 2003).

FoxP2 expression was assessed in neonatal and prepubescent mouse brain samples exposed to DVD deficiency and PEE. Its expression was not altered in DVD or DVD-PEE samples, even though there is evidence to suggest that DVD deficiency can regulate the expression of this transcript in BALB/c mouse embryos (Hawes et al 2015). On the other hand, PEE resulted in a significant downregulation of FoxP2 at birth. Additionally, males from the PEE group showed a significantly increased number of calls during a maternal

140 separation test at P7. However, P0 gene expression findings were not correlated with behavioural parameters, and animals underwent further behavioural phenotyping, which did not allow for FoxP2 expression assessment at P7. FoxP2 is a highly conserved gene across species (humans, non-human primates, rodents) which has been associated with speech in numerous studies. It was initially identified in a British family (KE family) with specific language impairments, which appeared to be inherited in an autosomal dominant fashion (Hurst et al 1990). More recently, mouse pups with heterozygous non-functional FoxP2 alleles were shown to have mild developmental delays and produced fewer ultrasonic calls (Fujita et al 2008a, Shu et al 2005). It is now known that FoxP2 is not a “language” gene but a transcription factor that affects the function of many genes and is involved, for instance, in the development of the lungs, heart and other organs (Fisher et al 2006), and is critical in DA system regulation (Murugan et al 2013). It would be useful for future studies to independently replicate the altered number of calls at P7 and immediately collect brain tissue for qRT-PCR analysis of FoxP2 expression levels.

Locomotor activity and sociability were assessed in prepubescent mice (P21). These behaviours remained unchanged in DVD-deficient animals, while PEE male mice showed increased locomotion and blunted sociability. There is no previous evidence on the effects of DVD deficiency on juvenile mouse behaviour, however, adult DVD-deficient offspring showed strain-dependent hyperactivity (no change in C57BL/6J) and unaffected sociability (Harms et al 2008a). In the case of PEE, prepubescent hyperactivity is consistent with previous findings (Sanchez Vega et al 2013), however, sociability had not been characterized using the PEE model described in this thesis. Therefore, the impairment found in this measure is a novel finding, which concurs with reports from studies in rats, showing disrupted social behaviours (although using different tasks) in a sexually dimorphic manner (Lugo et al 2003, Meyer et al 1986).

Shortly after behavioural testing at P21, the gene expression of candidates known to be relevant in neurodevelopmental disorders such as ASD and ADHD, as well as in the regulation of neurotransmitter systems such as glutamate and dopamine, was assessed in brain samples. The downregulation of FoxP2 found at P0 did not persist into prepubescence, which suggests that compensatory mechanisms may have taken place between birth and P21 to stabilize its expression levels.

141

FoxP2 is predicted to be a strong target of miR-9 (it has two 7mer-m8 sites in the proximate 3’UTR). miRNAs are small, nonprotein-coding RNAs that control the expression of target gene networks through silencing (Miranda et al 2010). A study in mammalian neural stem cells showed that ethanol exposure can supress several miRNAs, one of them miR-9 (Balaraman et al 2012, Sathyan et al 2007). MiR-9 is an evolutionary conserved molecule, that is sensitive to ethanol during development (Sathyan et al 2007, Tal et al 2012, Wang et al 2009) and in the adulthood (Pietrzykowski et al 2008). These findings were also validated in-vivo using a zebrafish model, with data showing ethanol exposure during gastrulation results in a significant alteration of miR-9 expression relative to control samples, which in turn lead to a significant suppression of miR-9 mRNA targets FoxP2 and FGFR-1. Considering that one of miR-9 roles is to regulate FoxP2 expression via suppression, these findings suggest that ethanol can specifically supress mRNAs that are also targeted by miR-9, and by doing so it uncouples the regulatory function of miR-9. FoxP2 is likely controlled by multiple miRNAs, which may compensate for the downregulation of miR-9. MiR-132 is a possible candidate that may be implicated in this compensatory mechanism, given that it is involved in the prevention of ectopic expression of FoxP2 by targeting its 3’UTR. Additionally, a study in zebrafish showed that embryonic exposure to 1% ethanol 24h post-fertilization significantly upregulated miR-132 (Soares et al 2012). Taking into account that these findings are in a model using different species and exposure levels, it would be valuable to test the effects of ethanol exposure on miR-9/132 expression in the present PEE mouse model, followed by a correlation with FoxP2 expression.

Darpp-32 was another transcript found to be dysregulated upon PEE as measured at P0 and P21, which is a key DA signalling regulator. Darpp-32 is expressed in the striatum and cortex and is co-expressed with nearly all FoxP2+ neurons (adult tissue, not in interneurons), as it was observed in neonatal brain sections (Chapter 5). One of its roles is to communicate signals in response to dopamine D1 receptor activation (Fienberg et al 1998). D1 receptors stimulate the phosphorylation of Darpp-32 on Thr-34 via adenylyl cyclase and protein kinase A, thereby converting Darpp-32 to an inhibitor of protein phosphatase-1 (PP-1). This decreases endogenous phosphatase activity and enhances phosphorylation of one of the major subunits of the NMDA receptor, NR1. This has been shown to decrease NMDAR sensitivity to ethanol and promote long-term alterations in glutamatergic transmission (Maldve et al 2002). In vitro studies have shown that ethanol is capable of mediating Darpp-32 phosphorylation, however, the underlying mechanism is

142 unclear, and the effects of prenatal administration of ethanol on Darpp-32 has not been fully described. Regarding functional outcomes, there is some evidence to show Darpp-32 is involved in pathways that may influence behaviour, such as locomotion. Drd1 activation has an excitatory effect, it promotes phosphorylation of Darpp-32 and has a movement enhancing effect in terms of behavioural outcomes (Scott et al 2005). In contrast, D2R activation has an inhibitory effect over Darpp-32 activity (Stoof et al 1981), thereby resulting in decreased locomotion.

TH is the rate-limiting enzyme in DA synthesis (Molinoff et al 1971). Its mRNA expression was not altered by DVD deficiency or PEE, but has a significant main effect of sex, whereby females exposed to either risk factor (i.e. not controls) showed significantly higher levels of TH than males. Even though it requires further validation, the overexpression of TH in females could occur as part of compensatory actions that may explain the normalized behavioural phenotype observed in DVD and PEE females. A study in drosophila showed a sexually dimorphic effect in the stress response, whereby pre- and post-synaptic dopaminergic neurons were differentially recruited into the stress response circuitry for males and females (Argue et al 2013). Considering TH is the rate-limiting enzyme for DA synthesis, it is plausible that its expression in response to prenatal insults is disrupted in a sexually dimorphic manner, which may in turn result in differing behavioural outcomes in males and females.

Alternatively, it is possible that males are more vulnerable to prenatal insults than females. Previous studies have shown that (acute) ethanol treatment resulted in significant changes in neurotrophin receptor levels in the hippocampus, septum, cerebral cortex and cerebellum and males were more affected than females (Moore et al 2004). Another study found an association between PEE and a decreased volume of sexually dimorphic nucleus of the preoptic area (SND-POA) of the hypothalamus in male rats, along with a significant reduction of neuronal cell size in this area. The size of the SND-POA is known to be strongly influenced by circulating androgens and possibly oestrogens during late postnatal/early postnatal life, which implies that alterations in the endogenous levels of sex steroids could have adverse long-term effects on this region.

Previous studies of PEE and DA signalling have reported that male PEE rats show differential responses to Drd1 agonists (Blanchard et al 1987, Hannigan et al 1990, Means et al 1984, Ulug et al 1983) and antagonists (Hannigan 1990), which suggests that PEE

143 may preferentially increase the activity of DA receptors in male rats. Increased Drd1 binding (Converse et al 2014), lower DA concentration and higher DOPAC/DA ratios (Boggan et al 1996) was found in males compared to females. Overall, males appear to be more vulnerable to the adverse effects on the DA system from prenatal ethanol exposure, which is consistent with human clinical studies showing that FASD boys were more likely to have the DA-related diagnosis of ADHD than FASD girls (Herman et al., 2008). The flow chart below (Fig 7.2) postulates a plausible mechanism underlying the effects of PEE on the expression of DA-related candidates, resulting in altered behaviour of male but not female offspring. This proposed mechanism integrates evidence discussed in this thesis, including dysregulation of FoxP2 and Darpp-32, with candidates that can be experimentally examined in future studies, such as epigenetic markers miR-9 and -132.

Figure 7.2. Postulated FoxP2/Darpp-32 regulation by PEE. PEE has been shown to alter the expression of miRNAs -9 and -132, which in turn can epigenetically modulate the expression of FoxP2. This transcription factor was downregulated in PEE males at birth, along with Darpp-32, which was also upregulated at P21. Darpp-32 is known to be co-expressed with FoxP2 in MSNs of the striatum, and to mediate DA signalling via Drd1, which can in turn affect DA-related behaviours. Male PEE showed disrupted communication, locomotion and sociability, while female behaviour was unaffected. TH expression at P21 was upregulated in females but not males, which could be part of compensatory mechanisms that account for the normalized behaviour compared to male mice.

144

7.4 Impact of DVD deficiency and PEE on adult outcomes

Behaviour was also evaluated in adult offspring, with tasks focusing on glutamate, dopamine and hippocampal-dependent behaviours. Locomotion is a simple but sensitive measure, which can be disrupted by dysregulations in various neurotransmitter systems, including glutamate and dopamine. Results showed that DVD deficiency did not alter activity levels, which is consistent with previous reports on C57BL/6J mice. In the case of PEE, hypoactivity was found in males across two separate cohorts, which agrees with previous reports, although it contradicts a previous study using the same PEE model, during which hyperactivity was encountered in PEE mice (Sanchez Vega et al 2013) (possible limitations discussed in Chapter 2). Moreover, there were no significant changes in locomotion of DVD-PEE animals, suggesting there is no interaction between Diet and Exposure on this measure.

Based on evidence in the literature, which suggests that the hippocampus is particularly vulnerable to the effects of prenatal exposures, hippocampal-dependent memory was evaluated in adult offspring using the APA task. DVD deficiency did not impact the ability of adult offspring to learn the task, nor did PEE or the combination of the factors. Previous studies had shown subtle alterations in spatial learning and memory as a result of DVD deficiency (Fernandes de Abreu et al 2010) and PEE (Sanchez Vega et al 2013). However, these studies used different tasks, and perhaps experimental discrepancies such as housing conditions (discussed in Chapter 3), may have interfered with the results found in this study. In addition to hippocampal learning and memory, preliminary data suggested regulation of glutamatergic signalling may be altered in the hippocampus of PEE offspring. Vglut2 mRNA expression was assessed in the adult hippocampus of all experimental groups, but only a downregulation in the PEE group was observed. While directionally inconsistent with previous studies where epigenetic regulation of this gene was disrupted by PEE (Zhang et al 2015), this suggests that glutamatergic signalling is vulnerable to the effects of PEE. To test whether the disruption in glutamate transporter expression (Vglut2) had a functional outcome on glutamatergic signalling, which could in turn explain the locomotion changes observed in PEE males, adult male offspring underwent a psychomimetic (MK-801)-induced challenge. During this test, the hyperlocomotive response to MK-801 was assessed, and there was no significant effect of DVD deficiency, PEE or their combination, compared to controls, which suggests that the

145 changes observed at baseline locomotion are likely being driven by a neurotransmitter system other than glutamate, such as GABA (discussed in Chapter 3).

Even though the hippocampus was of primary focus due to preliminary reports on behaviour and gene expression changes in this area, perhaps this region is not the most vulnerable to the effects of PEE. PEE was shown to disrupt the expression of candidate genes such as FoxP2 and Darpp-32, which are involved in DA signalling regulation and are highly expressed in the striatum, and are not expressed in the hippocampus (Brene et al 1994, Ferland et al 2003). Therefore, it is possible that the effects of PEE during early postnatal stages are more prominent on the striatum and not the hippocampus. Consequently, dopamine-mediated behaviours regulated by the striatum, such as motivation, social and goal-directed behaviours may be impaired and warrant further investigation in future studies.

There are a number of possible reasons that may explain the lack of behavioural alterations in mice exposed to both DVD deficiency and PEE. Gene expression and behavioural data from this thesis suggest that the effects of DVD deficiency and PEE become less apparent as offspring age, possibly as a result of compensatory effects taking place after the reintroduction of vitamin D at birth. This suggests that the time around birth may be a critical period during which DVD deficiency and PEE increase the offspring’s vulnerability, however, additional postnatal stress factor(s) may be necessary to induce potent, long-lasting effects at a molecular and behavioural level. There is epidemiological data to support this hypothesis. A study examining the effects of prenatal stress found that maternal experience of multiple stressful events during pregnancy was significantly associated with a greater incidence of mental health morbidity in children between 2 and 14 years compared to children whose mothers did not report stress during pregnancy (Robinson et al 2011).

On the other hand, it is possible that the duration of DVD deficiency and the dosage of ethanol during this particular window of development (GD0-8) do not have an interactive effect on postnatal behaviours, however, it is also plausible that should other phenotypes be assayed, an exacerbated phenotype may be observed in the two-hit exposure group. Further work is required to characterise other phenotypes (i.e. measures of depressive-like or anxiety-like behaviours) to more comprehensively assess the presence of lack thereof of interaction in this model.

146

7.5 Limitations and future directions

The evidence discussed in this thesis provided novel information regarding the effects of DVD deficiency, PEE and their combination, from which several experiments can be designed to further investigate the consequences of these exposures. RNA sequencing screening revealed that the combination of DVD-PEE resulted in the dysregulation of several genes involved in various cellular processes that can alter neurodevelopmental trajectories. These were preliminary results that served to confirm the biological plausibility of the effects of DVD deficiency and PEE on transcriptional regulation of neurodevelopmental processes. Therefore, the gene expression assessment of top candidates via qRT-PCR is the next logical step to validate the RNA-Seq findings.

In terms of DA-signalling regulation, it would be useful to evaluate the epigenetic regulation of dysregulated candidate genes, FoxP2 and Darpp-32, to test whether speculated miRNAs (-9 and -132) are indeed affected by PEE and take part in the aberrant expression of FoxP2. Considering FoxP2 and Darpp-32 are primarily expressed in MSNs of the striatum, the expression of these candidates should be evaluated in a cell- targeted manner rather than in whole cerebrum tissue, followed by quantification of corresponding protein expression in striatal sections. Furthermore, based on evidence suggesting that FoxP2/Darpp-32 are involved in DA signalling, future behavioural assessments could focus on DA-mediated behaviours, including locomotion, sociability and reward-seeking, at both juvenile and adult stages, aiming to correlate results with gene expression levels. Additionally, sex hormone expression and HPA axis regulation would be valuable to examine and correlate to behavioural findings, which could account for the sexually dimorphic effects of PEE reported in this thesis, identifying that males are more vulnerable to PEE than females.

Another direction for future studies could be to modify experimental conditions to address the postnatal neutralisation of DVD-PEE effects in the offspring. One option could include adding a postnatal stress factor, such as post-weaning isolation or social stress, followed by an assessment of ASD-related behaviours in juvenile and adult mice. Since DVD-PEE dysregulated the expression of several genes at birth, it can be speculated that the combination of factors transiently increased the offspring’s vulnerability to stressors, and the addition of a postnatal risk factor may exacerbate these effects, resulting in a more severe behavioural phenotype relevant to neurodevelopmental disorders. Another option

147 would be to prolong the vitamin D deficiency period until weaning to investigate whether the delayed reintroduction of vitamin D (compared to the ‘classic’ model used in this thesis) results in more prominent changes in terms of behaviour and gene expression. Dietary calcium supplementation should be considered to prevent confounds in behavioural outcomes driven by hypocalcaemia.

7.6 Conclusions

The research presented in this thesis demonstrates that, for the first time, a two-hit model of two plausible environmental risk factors, DVD deficiency and PEE, was established in mice. The results showed that the combined effect of these exposures has a transient impact on brain function, affecting the regulation of several genes involved in cell signalling, migration and death, and then dissipating throughout postnatal stages. DVD deficiency alone resulted in subtle molecular and neuroanatomical alterations in neonates, which were in line with findings previously reported in the literature regarding the role of vitamin D in cell proliferation, adhesion and apoptosis. PEE was associated with specific changes in gene expression and behaviour, where males appeared to be more vulnerable than females. These data suggest that even short-term, moderate exposure to ethanol can have a long-lasting impact on DA signalling and related behaviours, and lend further support to clinical studies associating PEE and neurodevelopmental disorders such as ASD and ADHD. Overall, the DVD-PEE mouse model provided valuable information regarding the timing and interaction of these two risk factors, highlighting that crucial cellular processes are vulnerable to subtle exposures and suggesting that their combination may increase the risk for neurodevelopmental disorders in the offspring, and for that reason, optimal vitamin D levels and abstinence form alcohol during pregnancy are recommended.

148

References

*Aarnoudse-Moens CSH, Weisglas-Kuperus N, van Goudoever JB, Oosterlaan J. 2009. Meta-analysis of neurobehavioral outcomes in very preterm and/or very low birth weight children. Pediatrics 124: 717 Abate P, Reyes-Guzmán AC, Hernández-Fonseca K, Méndez M. 2017. Prenatal ethanol exposure modifies locomotor activity and induces selective changes in Met-enk expression in adolescent rats. Neuropeptides 62: 45-56 Abbott CW, Kozanian OO, Kanaan J, Wendel KM, Huffman KJ. 2016. The impact of prenatal ethanol exposure on neuroanatomical and behavioral development in mice. Alcohol Clin Exp Res 40: 122-33 Abel EL. 1984. Prenatal effects of alcohol. J Alcohol Drug Depend 14: 1-10 Abel EL, Kruger ML, Friedl J. 1998. How do physicians define "light", "moderate", and "heavy" drinking? Alcohol Clin Exp Res 22: 979-84 Addolorato G, Capristo E, Greco AV, Stefanini GF, Gasbarrini G. 1997. Energy expenditure, substrate oxidation, and body composition in subjects with chronic alcoholism: new findings from metabolic assessment. Alcohol Clin Exp Res 21: 962- 7 Adelmant G, Begue A, Stehelin D, Laudet V. 1996. A functional Rev-erb alpha responsive element located in the human Rev-erb alpha promoter mediates a repressing activity. Proc Natl Acad Sci U S A 93: 3553-8 Aghajafari F, Nagulesapillai T, Ronksley PE, Tough SC, O’beirne M, Rabi DM. 2013. Association between maternal serum 25-hydroxyvitamin D level and pregnancy and neonatal outcomes: systematic review and meta-analysis of observational studies. BMJ : British Medical Journal 346 Ahlgren SC, Thakur V, Bronner-Fraser M. 2002. Sonic hedgehog rescues cranial neural crest from cell death induced by ethanol exposure. Proc Natl Acad Sci U S A 99: 10476 Alexianu ME, Robbins E, Carswell S, Appel SH. 1998. 1 alpha,25 dihydroxyvitamin D-3- dependent up-regulation of calcium-binding proteins in motoneuron cells. J Neurosci Res 51: 58-66 Allan AM, Chynoweth J, Tyler LA, Caldwell KK. 2003. A mouse model of prenatal ethanol exposure using a voluntary drinking paradigm. Alcohol Clin Exp Res 27: 2009-16

149

Almeras L, Eyles D, Benech P, Laffite D, Villard C, et al. 2007. Developmental vitamin D deficiency alters brain protein expression in the adult rat: implications for neuropsychiatric disorders. Proteomics 7: 769-80 Álvarez-Díaz S, Larriba MJ, López-Otín C, Muñoz A. 2010. Vitamin D: Proteases, protease inhibitors and cancer. Cell Cycle 9: 32-37 Ammassari-Teule M, Sgobio C, Biamonte F, Marrone C, Mercuri NB, Keller F. 2009. Reelin haploinsufficiency reduces the density of PV+ neurons in circumscribed regions of the striatum and selectively alters striatal-based behaviors. Psychopharmacology (Berl) 204: 511-21 Anders S, Pyl PT, Huber W. 2015. HTSeq--a Python framework to work with high- throughput sequencing data. Bioinformatics (Oxford, England) 31: 166-9 Anderson SM, Pierce RC. 2005. Cocaine-induced alterations in dopamine receptor signaling: Implications for reinforcement and reinstatement. Pharmacol Ther 106: 389-403 Annweiler C, Allali G, Allain P, Bridenbaugh S, Schott AM, et al. 2009. Vitamin D and cognitive performance in adults: a systematic review. Eur J Neurol 16: 1083-89 Anthony B, Zhou FC, Ogawa T, Goodlett CR, Ruiz J. 2008. Alcohol exposure alters cell cycle and apoptotic events during early neurulation. Alcohol Alcohol 43: 261-73 Argue KJ, Neckameyer WS. 2013. Sexually Dimorphic Recruitment of Dopamine Neurons into the Stress Response Circuitry. Behav Neurosci 127: 734-43 Astrom A, Tavakkol A, Pettersson U, Cromie M, Elder JT, Voorhees JJ. 1991. Molecular cloning of two human cellular retinoic acid-binding proteins (CRABP). Retinoic acid- induced expression of CRABP-II but not CRABP-I in adult human skin in vivo and in skin fibroblasts in vitro. The Journal of biological chemistry 266: 17662-6 Badea A, Nicholls PJ, Johnson GA, Wetsel WC. 2007. Neuroanatomical phenotypes in the reeler mouse. Neuroimage 34: 1363-74 Baggio S, Mussulini BH, de Oliveira DL, Zenki KC, Santos Da Silva E, Rico EP. 2017. Embryonic alcohol exposure promotes long-term effects on cerebral glutamate transport of adult zebrafish. Neurosci Lett 636: 265-69 Bailey CDC, Brien J, Reynolds J. 2001. Chronic prenatal ethanol exposure increases gaba sub(a) receptor subunit protein expression in the adult guinea pig cerebral cortex. J Neurosci 21: 4381-89 Balaraman S, Winzer-Serhan UH, Miranda RC. 2012. Opposing actions of ethanol and nicotine on microRNAs are mediated by nicotinic acetylcholine receptors in fetal cerebral cortical-derived neural progenitor cells. Alcohol Clin Exp Res 36: 1669-77

150

Balden R, Selvamani A, Sohrabji F. 2012. Vitamin D deficiency exacerbates experimental stroke injury and dysregulates ischemia-induced inflammation in adult rats. Endocrinol 153: 2420-35 Bancroft JD, Gamble M. 2008. Theory and practice of histological techniques Philadelphia, PA: Philadelphia, PA : Churchill Livingstone/Elsevier. Banerjee TD, Middleton F, Faraone SV. 2007. Environmental risk factors for attention- deficit hyperactivity disorder. Acta Paediatr 96: 1269-74 Barbazanges A, Vallee M, Mayo W, Day J, Simon H, et al. 1996. Early and later adoptions have different long-term effects on male rat offspring. J Neurosci 16: 7783 Barker DJ, Osmond C. 1986. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet 1: 1077-81 Barron S, Gilbertson R. 2005. Neonatal ethanol exposure but not neonatal cocaine selectively reduces specific isolation-induced vocalization waveforms in rats. Behav Genet 35: 93-102 Barsony J, Prufer K. 2002. Vitamin D receptor and retinoid X receptor interactions in motion. Vitamins and hormones 65: 345-76 Becker A, Eyles DW, McGrath JJ, Grecksch G. 2005. Transient prenatal vitamin D deficiency is associated with subtle alterations in learning and memory functions in adult rats. Behav Brain Res 161: 306-12 Becker HC, Becker HC, Randall CL, Randall CL. 1989. Effects of prenatal ethanol exposure in C57BL mice on locomotor activity and passive avoidance behavior. Psychopharmacology 97: 40-44 Bell R, Kimpel M, McClintick JN, N Strother W, Carr L, et al. 2009. Gene expression changes in the nucleus accumbens of alcohol-preferring rats following chronic ethanol consumption. Pharmacol Biochem Behav 94: 131-47 Berghofer-Hochheimer Y, Zurek C, Wolfl S, Hemmerich P, Munder T. 1998. L7 protein is a coregulator of vitamin D receptor-retinoid X receptor-mediated transactivation. J Cell Biochem 69: 1-12 Bergson C, Mrzljak L, Smiley JF, Pappy M, Levenson R, Goldman-Rakic PS. 1995. Regional, cellular, and subcellular variations in the distribution of D1 and D5 dopamine receptors in primate brain. J Neurosci 15: 7821-36 Bhutta AT, Cleves MA, Casey PH, Cradock MM, Anand KJS. 2002. Cognitive and Behavioral Outcomes of School-Aged Children Who Were Born Preterm: A Meta- analysis. JAMA 288: 728-37

151

Blanchard BA, Hannigan JH, Riley EP. 1987. Amphetamine-induced activity after fetal alcohol exposure and undernutrition in rats. Neurotoxicol Teratol 9: 113-9 Bodnar LM, Simhan HN, Powers RW, Frank MP, Cooperstein E, Roberts JM. 2007. High prevalence of vitamin D insufficiency in black and white pregnant women residing in the northern United States and their neonates. J Nutr 137: 447-52 Boggan WO, Xu W, Shepherd CL, Middaugh LD. 1996. Effects of prenatal ethanol exposure on dopamine systems in C57BL/6J mice. Neurotoxicol Teratol 18: 41-8 Bond NW, Digiusto EL. 1977. Prenatal alcohol consumption and open-field behavior in rats - effects of age at time of testing. Psychopharmacology 52: 311-12 Borowicz KK, Morawska M, Furmanek-Karwowska K, Luszczki JJ, Czuczwar SJ. 2007. Cholecalciferol enhances the anticonvulsant effect of conventional antiepileptic drugs in the mouse model of maximal electroshock. European journal of pharmacology 573: 111-5 Branchi I, Santucci D, Alleva E. 2001. Ultrasonic vocalisation emitted by infant rodents: a tool for assessment of neurobehavioural development. Behav Brain Res 125: 49-56 Brene S, Lindefors N, Ehrlich M, Taubes T, Horiuchi A, et al. 1994. Expression of mRNAs encoding ARPP-16/19, ARPP-21, and DARPP-32 in human brain tissue. J Neurosci 14: 985 Brewer LD, Thibault V, Chen KC, Langub MC, Landfield PW, Porter NM. 2001. Vitamin D hormone confers neuroprotection in parallel with downregulation of L-type calcium channel expression in hippocampal neurons. J Neurosci 21: 98-108 Brolese G, Lunardi P, Broetto N, Engelke DS, Lírio F, et al. 2014. Moderate prenatal alcohol exposure alters behavior and neuroglial parameters in adolescent rats. Behav Brain Res 269: 175-84 Brown AJ, Zhong M, Finch J, Ritter C, McCracken R, et al. 1996. Rat calcium-sensing receptor is regulated by vitamin D but not by calcium. The American journal of physiology 270: F454-60 Brown G, Hughes PJ, Michell RH. 2003. Cell differentiation and proliferation— simultaneous but independent? Exp Cell Res 291: 282-88 Brown RT, Coles CD, Smith IE, Platzman KA, Silverstein J, et al. 1991. Effects of Prenatal Alcohol Exposure at School Age .2. Attention and Behavior. Neurotoxicol Teratol 13: 369-76 Bulfone A, Smiga SM, Shimamura K, Peterson A, Puelles L, Rubenstein JLR. 1995. T- Brain-1: A homolog of whose expression defines molecularly distinct domains within the cerebral cortex. Neuron 15: 63-78

152

Burne TH, Becker A, Brown J, Eyles DW, Mackay-Sim A, McGrath JJ. 2004a. Transient prenatal Vitamin D deficiency is associated with hyperlocomotion in adult rats. Behav Brain Res 154: 549-55 Burne TH, O'Loan J, McGrath JJ, Eyles DW. 2006. Hyperlocomotion associated with transient prenatal vitamin D deficiency is ameliorated by acute restraint. Behav Brain Res 174: 119-24 Burne TH, O'Loan J, Splatt K, Alexander S, McGrath JJ, Eyles DW. 2011a. Developmental vitamin D (DVD) deficiency alters pup-retrieval but not isolation-induced pup ultrasonic vocalizations in the rat. Physiol Behav 102: 201-4 Burne THJ, Becker A, Brown J, Eyles DW, Mackay-Sim A, McGrath JJ. 2004b. Transient prenatal Vitamin D deficiency is associated with hyperlocomotion in adult rats. Behav Brain Res 154: 549-55 Burne THJ, McGrath JJ, Eyles DW, Mackay-Sim A. 2005. Behavioural characterization of Vitamin D receptor knockout mice. Behav Brain Res 157: 299-308 Burne THJ, O'Loan J, Splatt K, Alexander S, McGrath JJ, Eyles DW. 2011b. Developmental vitamin D (DVD) deficiency alters pup-retrieval but not isolation- induced pup ultrasonic vocalizations in the rat. Physiol Behav 102: 201-04 Callinan S, Room R. 2012. Alcohol consumption during pregnancy: results from the 2010 national drug strategy household survey. Found Alc Res Educ Camargo Moreno M, Mooney SM, Middleton FA. 2017. Heterogeneity of p53 dependent genomic responses following ethanol exposure in a developmental mouse model of fetal alcohol spectrum disorder. PLoS One 12: e0180873 Campbell P, Reep RL, Stoll ML, Ophir AG, Phelps SM. 2009. Conservation and diversity of Foxp2 expression in muroid rodents: Functional implications. J Comp Neurol 512: 84-100 Canaff L, Hendy GN. 2002. Human calcium-sensing receptor gene. Vitamin D response elements in promoters P1 and P2 confer transcriptional responsiveness to 1,25- dihydroxyvitamin D. The Journal of biological chemistry 277: 30337-50 Cannell JJ. 2008. Autism and vitamin D. Med Hypotheses 70: 750-59 Cao G, Gu Z, Ren Y, Shu L, Tao C, et al. 2009. Parathyroid hormone contributes to regulating milk calcium content and modulates neonatal bone formation cooperatively with calcium. Endocrinol 150: 561-69 Carden SE, Barr GA, Hofer MA. 1991. Differential effects of specific opioid receptor agonists on rat pup isolation calls. Brain research. Developmental brain research 62: 17-22

153

Cartwright MM, Tessmer L, Smith SM. 1998. Ethanol-induced neural crest apoptosis is coincident with their endogenous death, but is mechanistically distinct. Alcoholism (NY) 22: 142-49 Chen CP, Kuhn P, Advis JP, Sarkar DK. 2006. Prenatal ethanol exposure alters the expression of period genes governing the circadian function of β-endorphin neurons in the hypothalamus. J Neurochem 97: 1026-33 Chen L, Wilson R, Bennett E, Zosky GR. 2016. Identification of vitamin D sensitive pathways during lung development. Respir Res 17: 47 Chen TC, Chimeh F, Lu Z, Mathieu J, Person KS, et al. 2007. Factors that influence the cutaneous synthesis and dietary sources of vitamin D. Arch. Biochem. Biophys. 460: 213-17 Cheng XR, Yang Y, Zhou WX, Zhang YX. 2011. Expression of VGLUTs contributes to degeneration and acquisition of learning and memory. Neurobiol Learn Mem 95: 361-75 Chiu YC, Li MY, Liu YH, Ding JY, Yu JY, Wang TW. 2014. Foxp2 regulates neuronal differentiation and neuronal subtype specification. Dev Neurobiol 74: 723-38 Christakos S, Dhawan P, Benn B, Porta A, Zhong Y, et al. 2007. Vitamin D: Molecular mechanism of action. pp. 340-48 Cimadevilla JM, Wesierska M, Fenton AA, Bures J. 2001. Inactivating one hippocampus impairs avoidance of a stable room-defined place during dissociation of arena cues from room cues by rotation of the arena. Proc Natl Acad Sci U S A 98: 3531-6 Coles CD, Goldstein FC, Lynch ME, Chen XC, Kable JA, et al. 2011. Memory and brain volume in adults prenatally exposed to alcohol. Brain Cognition 75: 67-77 Colvin L, Payne J, Parsons D, Kurinczuk JJ, Bower C. 2007. Alcohol consumption during pregnancy in nonindigenous west Australian women. Alcohol Clin Exp Res 31: 276- 84 Committee on Understanding Premature B, Assuring Healthy O, Behrman RE, Butler AS. 2006. Preterm Birth: Causes, consequences, and prevention. National Academies Press. Consiglio M, Destefanis M, Morena D, Foglizzo V, Forneris M, et al. 2015. The vitamin D receptor inhibits the respiratory chain, contributing to the metabolic switch that is essential for cancer cell proliferation. PLoS One 9: e115816 Converse AK, Moore CF, Holden JE, Ahlers EO, Moirano JM, et al. 2014. Moderate level prenatal alcohol exposure induces sex differences in dopamine D(1) receptor binding in adult rhesus monkeys. Alcohol Clin Exp Res 38: 2934-43

154

Cooper D, Rudeen K. 1988. Alterations in regional catecholamine content and turnover in the male rat brain in response to in utero ethanol exposure. Alcohol Clin Exp Res 12: 282-85 Corominas R, Yang X, Lin G, Kang S, Shen Y, et al. 2014. Protein interaction network of alternatively spliced isoforms from brain links genetic risk factors for autism. Nat. Commun. 5 Crawley JN. 2007. Mouse behavioral assays relevant to the symptoms of autism. Brain pathology (Zurich, Switzerland) 17: 448-59 Crawley JN, Paylor R. 1997. A proposed test battery and constellations of specific behavioral paradigms to investigate the behavioral phenotypes of transgenic and knockout mice. Horm Behav 31: 197-211 Cui X, Pelekanos M, Burne THJ, McGrath JJ, Eyles DW. 2010. Maternal vitamin D deficiency alters the expression of genes involved in dopamine specification in the developing rat mesencephalon. Neurosci Lett 486: 220-23 Cullen CL, Burne THJ, Lavidis NA, Moritz KM. 2013. Low dose prenatal ethanol exposure induces anxiety-like behaviour and alters dendritic morphology in the basolateral amygdala of rat offspring. PLoS One 8: e54924 Dallaspezia S, Locatelli C, Lorenzi C, Pirovano A, Colombo C, Benedetti F. 2016. Sleep homeostatic pressure and PER3 VNTR gene polymorphism influence antidepressant response to sleep deprivation in bipolar depression. Journal of affective disorders 192: 64-9 Dalvie S, King A, Fein G, Ramesar R, Stein DJ. 2016. Possible involvement of the circadian pathway in alcohol use disorder in a South African adolescent cohort. Met Brain Dis 31: 75-80 Daly RM, Gagnon C, Lu ZX, Magliano DJ, Dunstan DW, et al. 2012. Prevalence of vitamin D deficiency and its determinants in Australian adults aged 25 years and older: a national, population-based study. Clin Endocrinol 77: 26-35 Davies DL, Smith DE. 1981. A Golgi study of mouse hippocampal CA1 pyramidal neurons following perinatal ethanol exposure. Neurosci Lett 26: 49-54 De-Regil LM, Palacios C, Lombardo LK, Peña-Rosas JP. 2016. Vitamin D supplementation for women during pregnancy. Sao Paulo Med J 134: 274 Deluca H. 2004. Overview of general physiologic features and functions of vitamin D. Am J Clin Nutr 80: 1689S-96S Deriziotis P, Fisher SE. 2013. Neurogenomics of speech and language disorders: the road ahead. Genome biology 14: 204

155

Detering N, Collins RM, Hawkins RL, Ozand PT, Karahasan A. 1980. Comparative effects of ethanol and malnutrition on the development of catecholamine neurons: Changes in neurotransmitter levels. J Neurochem 34: 1587-93 Downing C, Flink S, Florez-McClure ML, Johnson TE, Tabakoff B, Kechris KJ. 2012. Gene expression changes in C57BL/6J and DBA/2J mice following prenatal alcohol exposure. Alcohol Clin Exp Res 36: 1519-29 Druse MJ, Tajuddin N, Kuo A, Connerty M. 1990. Effects of in utero ethanol exposure on the developing dopaminergic system in rats. J Neurosci Res 27: 233-40 Duester G. 1998. Alcohol dehydrogenase as a critical mediator of retinoic acid synthesis from vitamin A in the mouse embryo. J Nutr 128: 459s-62s Dursun E, Gezen-Ak D, Yilmazer S. 2011. A novel perspective for Alzheimer's disease: vitamin D receptor suppression by amyloid-beta and preventing the amyloid-beta induced alterations by vitamin D in cortical neurons. Journal of Alzheimer's disease : JAD 23: 207-19 Earnest DJ, Chen WJ, West JR. 2001. Developmental alcohol and circadian clock function. Alcohol Res Health 25: 136-40 Eliasen M, Tolstrup JS, Nybo Andersen A-M, Grønbaek M, Olsen J, Strandberg-Larsen K. 2010. Prenatal alcohol exposure and autistic spectrum disorders--a population- based prospective study of 80,552 children and their mothers. Int J Epidemiol 39: 1074-81 Elsabbagh M, Divan G, Koh YJ, Kim YS, Kauchali S, et al. 2012. Global prevalence of autism and other pervasive developmental disorders. Autism research : official journal of the International Society for Autism Research 5: 160-79 Enard W, Gehre S, Hammerschmidt K, Hölter SM, Blass T, et al. 2009. A humanized version of affects cortico-basal ganglia circuits in mice. Cell 137: 961-71 Evans SJ, Choudary PV, Neal CR, Li JZ, Vawter MP, et al. 2004. Dysregulation of the fibroblast growth factor system in major depression. Proc Natl Acad Sci U S A 101: 15506-11 Eyles D, Almeras L, Benech P, Patatian A, Mackay-Sim A, et al. 2007. Developmental vitamin D deficiency alters the expression of genes encoding mitochondrial, cytoskeletal and synaptic proteins in the adult rat brain. J Steroid Biochem Mol Biol 103: 538-45 Eyles D, Brown J, Mackay-Sim A, McGrath J, Feron F. 2003. Vitamin D3 and brain development. Neurosci 118: 641-53

156

Eyles DW, Feron F, Cui X, Kesby JP, Harms LH, et al. 2009. Developmental vitamin D deficiency causes abnormal brain development. Psychoneuroendocrino 34 Suppl 1: S247-57 Eyles DW, Smith S, Kinobe R, Hewison M, McGrath JJ. 2005. Distribution of the Vitamin D receptor and 1α-hydroxylase in human brain. J Chem Neuroanat 29: 21-30 Farrell WJ, Alberts JR. 2002. Maternal responsiveness to infant Norway rat (Rattus norvegicus) ultrasonic vocalizations during the maternal behavior cycle and after steroid and experiential induction regimens. Journal of comparative psychology (Washington, D.C. : 1983) 116: 286-96 Ferland RJ, Cherry TJ, Preware PO, Morrisey EE, Walsh CA. 2003. Characterization of Foxp2 and Foxp1 mRNA and protein in the developing and mature brain. J Comp Neurol 460: 266-79 Fernandes de Abreu DA, Nivet E, Baril N, Khrestchatisky M, Roman F, Féron F. 2010. Developmental vitamin D deficiency alters learning in C57Bl/6J mice. Behav Brain Res 208: 603-08 Fernell E, Barnevik‐Olsson M, Bågenholm G, Gillberg C, Gustafsson S, Sääf M. 2010. Serum levels of 25‐hydroxyvitamin D in mothers of Swedish and of Somali origin who have children with and without autism. Acta Paediatr 99: 743-47 Féron F, Burne THJ, Brown J, Smith E, McGrath JJ, et al. 2005. Developmental Vitamin D 3 deficiency alters the adult rat brain. Brain Res Bull 65: 141-48 Fienberg AA, Hiroi N, Mermelstein PG, Song W, Snyder GL, et al. 1998. DARPP-32: regulator of the efficacy of dopaminergic neurotransmission. Science 281: 838-42 Fisher SE, Marcus GF. 2006. The eloquent ape: genes, brains and the of language. Nat Rev Genet 7: 9-20 Fleet JC. 2008. Molecular actions of vitamin D contributing to cancer prevention. Mol Aspects Med 29: 388-96 Fleet JC, DeSmet M, Johnson R, Li Y. 2012. Vitamin D and cancer: A review of molecular mechanisms. The Biochemical journal 441: 61-76 Fleming AS, O’Day DH, Kraemer GW. 1999. Neurobiology of mother–infant interactions: experience and central nervous system plasticity across development and generations. Neurosci Biobehav Rev 23: 673-85 Fonseca BD, Zakaria C, Jia JJ, Graber TE, Svitkin Y, et al. 2015. La-related Protein 1 (LARP1) represses Terminal Oligopyrimidine (TOP) mRNA translation downstream of mTOR Complex 1 (mTORC1). The Journal of biological chemistry 290: 15996- 6020

157

Fox WM. 1965. Reflex-ontogeny and behavioural development of the mouse. Anim Behav 13: 234-41,IN5 Fremeau RT, Kam K, Qureshi T, Johnson J, Copenhagen DR, et al. 2004. Vesicular glutamate transporters 1 and 2 target to functionally distinct synaptic release sites. Science 304: 1815-19 Fujita E, Tanabe Y, Shiota A, Ueda M, Suwa K, et al. 2008a. Ultrasonic vocalization impairment of Foxp2 (R552H) knockin mice related to speech-language disorder and abnormality of Purkinje cells. Proc Natl Acad Sci U S A 105: 3117-22 Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, et al. 2008b. Tight junction proteins claudin-2 and -12 are critical for vitamin d-dependent ca(2+) absorption between enterocytes. Molecular and cellular biology 19: 1912-21 Fuller PJ. 1991. The steroid receptor superfamily: mechanisms of diversity. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 5: 3092-9 Fumagalli S, Thomas G. 2011. The role of p53 in ribosomopathies. Seminars in hematology 48: 97-105 Gaigg SB, Bowler DM. 2007. Differential fear conditioning in Asperger's syndrome: implications for an amygdala theory of autism. Neuropsychol 45: 2125-34 Ganu RS, Harris RA, Collins K, Aagaard KM. 2012. Early origins of adult disease: approaches for investigating the programmable epigenome in humans, nonhuman primates, and rodents. ILAR journal 53: 306-21 Gardener H, Spiegelman D, Buka SL. 2009. Prenatal risk factors for autism: comprehensive meta-analysis. BJPsych Int 195: 7-14 Gariépy J-L, Rodriguiz RM, Jones BC. 2002. Handling, genetic and housing effects on the mouse stress system, dopamine function, and behavior. Pharmacol Biochem Behav 73: 7-17 Gaughran F, Payne J, Sedgwick PM, Cotter D, Berry M. 2006. Hippocampal FGF-2 and FGFR1 mRNA expression in major depression, schizophrenia and bipolar disorder. Brain Res Bull 70: 221-27 Gauthier J, Joober R, Mottron L, Laurent S, Fuchs M, et al. 2003. Mutation screening of FOXP2 in individuals diagnosed with autistic disorder. Am J Med Genet A 118: 172- 75 Gerardino L, Papaleo P, Flex A, Gaetani E, Fioroni G, et al. 2006. Coagulation factor XIII Val34Leu gene polymorphism and Alzheimer's disease. Neurological research 28: 807-9

158

Gerfen CR, Engber TM, Mahan LC, Susel Z, Chase TN, et al. 1990. D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neurons. Science 250: 1429-32 Gezen-Ak D, Dursun E, Yilmazer S. 2011. The effects of vitamin D receptor silencing on the expression of LVSCC-A1C and LVSCC-A1D and the release of NGF in cortical neurons. PLoS One 6: e17553 Ghazalpour A, Bennett B, Petyuk VA, Orozco L, Hagopian R, et al. 2011. Comparative analysis of proteome and transcriptome variation in mouse. Plos Genet 7: e1001393 Giglia RC, Binns CW. 2007. Patterns of alcohol intake of pregnant and lactating women in Perth, Australia. Drug Alcohol Rev 26: 493-500 Ginde AA, Liu MC, Camargo CA, Jr. 2009. Demographic differences and trends of vitamin D insufficiency in the US population, 1988-2004. Arch Intern Med 169: 626-32 Girault V, Gilard V, Marguet F, Lesueur C, Hauchecorne M, et al. 2017. Prenatal alcohol exposure impairs autophagy in neonatal brain cortical microvessels. Cell Death Dis 8: e2610 Gong S, Zheng C, Doughty ML, Losos K, Didkovsky N, et al. 2003. A gene expression atlas of the central nervous system based on bacterial artificial . Nature 425: 917 Grant WB, Cannell JJ. 2013. Autism prevalence in the United States with respect to solar UV-B doses: An ecological study. Dermatoendocrinol 5: 159-64 Grant WB, Soles CM. 2009. Epidemiologic evidence supporting the role of maternal vitamin D deficiency as a risk factor for the development of infantile autism. Dermatoendocrinol 1: 223 Green ML, Singh AV, Zhang Y, Nemeth KA, Sulik KK, Knudsen TB. 2007. Reprogramming of genetic networks during initiation of the Fetal Alcohol Syndrome. Dev Dyn 236: 613-31 Greengard P, Allen PB, Nairn AC. 1999. Beyond the dopamine receptor: the DARPP- 32/protein phosphatase-1 cascade. Neuron 23: 435-47 Groves NJ, Kesby JP, Eyles DW, McGrath JJ, Mackay-Sim A, Burne THJ. 2013. Adult vitamin D deficiency leads to behavioural and brain neurochemical alterations in C57BL/6J and BALB/c mice. Behav Brain Res 241: 120-31 Guerri C. 1998. Neuroanatomical and neurophysiological mechanisms involved in central nervous system dysfunctions induced by prenatal alcohol exposure. Alcohol Clin Exp Res 22: 304-12

159

Guerri C. 2002. Mechanisms Involved in Central Nervous System Dysfunctions Induced by Prenatal Ethanol Exposure. Neurotox Res 4: 327-35 Gutala R, Wang J, Kadapakkam S, Hwang Y, Ticku M, Li MD. 2004. Microarray analysis of ethanol-treated cortical neurons reveals disruption of genes related to the ubiquitin- proteasome pathway and protein synthesis. Alcohol Clin Exp Res 28: 1779-88 Gutierrez-Monreal MA, Cuevas-Diaz Duran R, Moreno-Cuevas JE, Scott SP. 2014. A role for 1alpha,25-dihydroxyvitamin d3 in the expression of circadian genes. Journal of biological rhythms 29: 384-8 Hale RL, Randall CL, Becker HC, Middaugh LD. 1992. The effect of prenatal ethanol exposure on scentmarking in the C57BL/6J and C3H/He mouse strains. Alcohol 9: 287-92 Hamilton DA, Barto D, Rodriguez CI, Magcalas CM, Fink BC, et al. 2014. Effects of moderate prenatal ethanol exposure and age on social behavior, spatial response perseveration errors and motor behavior. Behav Brain Res 269: 44-54 Hannigan JH. 1990. The ontogeny of SCH 23390-induced catalepsy in male and female rats exposed to ethanol in utero. Alcohol 7: 11-6 Hannigan JH, Blanchard BA, Horner MP, Riley EP, Pilati ML. 1990. Apomorphine-induced motor behavior in rats exposed prenatally to alcohol. Neurotoxicol Teratol 12: 79-84 Hanson MA, Gluckman PD. 2014. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiological reviews 94: 1027-76 Happé F, Ehlers S, Fletcher P, Frith U, Johansson M, et al. 1996. 'Theory of mind' in the brain. Evidence from a PET scan study of Asperger syndrome. Neuroreport 8: 197 Happé F, Ronald A. 2008. The ‘Fractionable Autism Triad’: A review of evidence from behavioural, genetic, cognitive and neural research. Neuropsychol Rev 18: 287-304 Harding HP, Lazar MA. 1993. The orphan receptor Rev-ErbA alpha activates transcription via a novel response element. Molecular and cellular biology 13: 3113-21 Harding HP, Lazar MA. 1995. The monomer-binding orphan receptor Rev-Erb represses transcription as a dimer on a novel direct repeat. Molecular and cellular biology 15: 4791-802 Harms LR. 2011. Brain development and behaviour in developmentally vitamin D-deficient mice. The University of Queensland, Queensland Brain Institute Harms LR, Cowin G, Eyles DW, Kurniawan ND, McGrath JJ, Burne TH. 2012a. Neuroanatomy and psychomimetic-induced locomotion in C57BL/6J and 129/X1SvJ mice exposed to developmental vitamin D deficiency. Behav Brain Res 230: 125-31

160

Harms LR, Eyles DW, McGrath JJ, Mackay-Sim A, Burne TH. 2008a. Developmental vitamin D deficiency alters adult behaviour in 129/SvJ and C57BL/6J mice. Behav Brain Res 187: 343-50 Harms LR, Eyles DW, McGrath JJ, Mackay-Sim A, Burne THJ. 2008b. Developmental vitamin D deficiency alters adult behaviour in 129/SvJ and C57BL/6J mice. Behav Brain Res 187: 343-50 Harms LR, Turner KM, Eyles DW, Young JW, McGrath JJ, Burne TH. 2012b. Attentional processing in C57BL/6J mice exposed to developmental vitamin D deficiency. PLoS One 7: e35896 Hashimoto H, Saito TR, Furudate S-I, Takahashi KW. 2001. Prolactin levels and maternal behavior induced by ultrasonic vocalizations of the rat pup. Exp Anim 50: 307-12 Hashimoto-Torii K, Kawasawa YI, Kuhn A, Rakic P. 2011. Combined transcriptome analysis of fetal human and mouse cerebral cortex exposed to alcohol. Proc Natl Acad Sci U S A 108: 4212 Hawes JE, Tesic D, Whitehouse AJ, Zosky GR, Smith JT, Wyrwoll CS. 2015. Maternal vitamin D deficiency alters fetal brain development in the BALB/c mouse. Behav Brain Res 286: 192-200 Hebert KJ, Miller LL, Joinson CJ. 2010. Association of autistic spectrum disorder with season of birth and conception in a UK cohort. Autism research : official journal of the International Society for Autism Research 3: 185-90 Heindel JJ, Skalla LA, Joubert BR, Dilworth CH, Gray KA. 2017. Review of developmental origins of health and disease publications in environmental epidemiology. Reprod Toxicol 68: 34-48 Hellemans KGC, Verma P, Yoon E, Yu WK, Young AH, Weinberg J. 2010. Prenatal alcohol exposure and chronic mild stress differentially alter depressive‐ and anxiety‐ like behaviors in male and female offspring. Alcohol Clin Exp Res 34: 633-45 Hemmerich P, von Mikecz A, Neumann F, Sozeri O, Wolff-Vorbeck G, et al. 1993. Structural and functional properties of ribosomal protein L7 from humans and rodents. Nucleic acids research 21: 223-31 Hendy GN, Canaff L. 2016. Calcium-Sensing Receptor Gene: Regulation of Expression. Front Physiol 7: 394 Herrmann M, King K, Weitzman M. 2008. Prenatal tobacco smoke and postnatal secondhand smoke exposure and child neurodevelopment. Curr Opin Pediatr 20: 184-90

161

Hevner RF, Shi L, Justice N, Hsueh Y-P, Sheng M, et al. 2001. Tbr1 regulates differentiation of the preplate and layer 6. Neuron 29: 353-66 Hewson QC, Lovat PE, Pearson AD, Redfern CP. 2002. Retinoid signalling and gene expression in neuroblastoma cells: RXR agonist and antagonist effects on CRABP- II and RARbeta expression. J Cell Biochem 87: 284-91 Holick MF. 1987. Photosynthesis of vitamin D in the skin: Effect of environmental and life- style variables. Federation Proceedings 46: 1876-82 Holick MF. 1995. Environmental factors that tnfluence the cutaneous production of vitamin D. Am J Clin Nutr 61: S638-S45 Holick MF. 2003. Vitamin D: A millenium perspective. Journal Cell Biochem 88: 296-307 Holick MF. 2006. Resurrection of vitamin D deficiency and rickets. J Clin Investig 116: 2062-72 Hong CT, Wong CS, Ma HP, Wu D, Huang YH, et al. 2015. PERIOD3 polymorphism is associated with sleep quality recovery after a mild traumatic brain injury. Journal of the neurological sciences 358: 385-9 Horng S, Kreiman G, Ellsworth C, Page D, Blank M, et al. 2009. Differential gene expression in the developing lateral geniculate nucleus and medial geniculate nucleus reveals novel roles for Zic4 and Foxp2 in visual and auditory pathway development. J Neurosci 29: 13672 Hughes C, Russell J, Robbins TW. 1994. Evidence for executive dysfunction in autism. Neuropsychol 32: 477-92 Hurst JA, Baraitser M, Auger E, Graham F, Norell S. 1990. An extended family with a dominantly inherited speech disorder. Developmental medicine and child neurology 32: 352-5 Ibi M, Sawada H, Nakanishi M, Kume T, Katsuki H, et al. 2001. Protective effects of 1 alpha,25-(OH)(2)D-3 against the neurotoxicity of glutamate and reactive oxygen species in mesencephalic culture. Neuropharmacol 40: 761-71 Idrus NM MK, Happer JP, Thomas JD. . 2012. Vitamin D attenuates overactivity associated with developmental alcohol exposure in rats. Presented at Int Soc Biomed Res Alc, Sapporo, Japan Igarashi M, Nagata A, Toh H, Urade Y, Hayaishi O. 1992. Structural organization of the gene for prostaglandin D synthase in the rat brain. Proc Natl Acad Sci U S A 89: 5376-80

162

Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vöckler J, et al. 1999. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science 283: 70-74 Insel TR, Harbaugh CR. 1989. Central administration of corticotropin releasing factor alters rat pup isolation calls. Pharmacol Biochem Behav 32: 197-201 Ivins JK, Litwack ED, Kumbasar A, Stipp CS, Lander AD. 1997. Cerebroglycan, a developmentally regulated cell-surface heparan sulfate proteoglycan, is expressed on developing axons and growth cones. Developmental biology 184: 320-32 Jiang P, Zhang WY, Li HD, Cai HL, Liu YP, Chen LY. 2013. Stress and vitamin D: Altered vitamin D metabolism in both the hippocampus and myocardium of chronic unpredictable mild stress exposed rats. Psychoneuroendocrino 38: 2091-98 Jing H, Li Y. 2004. Effects of ethanol on mouse embryonic brain development and heat shock protein 73 expression. Toxicology in vitro : an international journal published in association with BIBRA 18: 601-7 Jones KL, Smith DW. 1973. Recognition of Fetal Alcohol Syndrome in Early Infancy. Lancet 2: 999-1001 Joyce MP, Carden SE. 1999. The effects of 8-OH-DPAT and (+/-)-pindolol on isolation- induced ultrasonic vocalizations in 3-, 10-, and 14-day-old rats. Dev Psychobiol 34: 109-17 Kalueff AV, Lou YR, Laaksi L, Tuohimaa P. 2004. Increased anxiety in mice lacking vitamin D receptor gene. Neuroreport 15: 1271-74 Kalueff AV, Minasyan A, Keisala T, Kuuslahti M, Miettinen S, Tuohimaa P. 2006a. Increased severity of chemically induced seizures in mice with partially deleted Vitamin D receptor gene. Neurosci Lett 394: 69-73 Kalueff AV, Minasyan A, Keisala T, Kuuslahti M, Miettinen S, Tuohimaa P. 2006b. The vitamin D neuroendocrine system as a target for novel neurotropic drugs. CNS & neurological disorders drug targets 5: 363-71 Kalueff AV, Minasyan A, Tuohimaa P. 2005. Anticonvulsant effects of 1,25- dihydroxyvitamin D in chemically induced seizures in mice. Brain Res Bull 67: 156- 60 Kaluzna-Czaplinska J, Socha E, Rynkowski J. 2010. Determination of homovanillic acid and vanillylmandelic acid in urine of autistic children by gas chromatography/mass spectrometry. Medical science monitor : international medical journal of experimental and clinical research 16: Cr445-50

163

Kaminen-Ahola N, Ahola A, Flatscher-Bader T, Wilkins SJ, Anderson GJ, et al. 2010a. Postnatal growth restriction and gene expression changes in a mouse model of fetal alcohol syndrome. Birth Defects Res A Clin Mol Teratol 88: 818-26 Kaminen-Ahola N, Ahola A, Maga M, Mallitt KA, Fahey P, et al. 2010b. Maternal ethanol consumption alters the epigenotype and the phenotype of offspring in a mouse model. Plos Genet 6: e1000811 Kang S, Jeong H, Baek JH, Lee SJ, Han SH, et al. 2016. PiB-PET imaging-based serum proteome profiles predict Mild Cognitive Impairment and Alzheimer's Disease. Journal of Alzheimer's disease : JAD 53: 1563-76 Kebabian JW, Calne DB. 1979. Multiple receptors for dopamine. Nature 277: 93-6 Kehoe P, Callahan M, Daigle A, Mallinson K, Brudzynski S. 2001. The effect of stimulation on rat pup ultrasonic vocalizations. Dev Psychobiol 38: 92-100 Kelly SJ, Dillingham RR. 1994. Sexually dimorphic effects of perinatal alcohol exposure on social interactions and amygdala DNA and DOPAC concentrations. Neurotoxicol Teratol 16: 377-84 Kelly SJ, Goodlett CR, Hannigan JH. 2009. Animal models of Fetal Alcohol Spectrum Disorders: Impact of the social environment. Dev Disabil Res Rev 15: 200-08 Kesby JP, Burne THJ, McGrath JJ, Eyles DW. 2006. Developmental vitamin D deficiency alters MK 801-induced hyperlocomotion in the adult rat: An animal model of schizophrenia. Biol Psychiatry 60: 591-96 Kesby JP, Cui X, Ko P, McGrath JJ, Burne THJ, Eyles DW. 2009. Developmental vitamin D deficiency alters dopamine turnover in neonatal rat forebrain. Neurosci Lett 461: 155-58 Kesby JP, Cui X, O'Loan J, McGrath JJ, Burne TH, Eyles DW. 2010. Developmental vitamin D deficiency alters dopamine-mediated behaviors and dopamine transporter function in adult female rats. Psychopharmacology (Berl) 208: 159-68 Kesby JP, O'Loan JC, Alexander S, Deng C, Huang XF, et al. 2012. Developmental vitamin D deficiency alters MK-801-induced behaviours in adult offspring. Psychopharmacology 220: 455-63 Kesby JP, Turner KM, Alexander S, Eyles DW, McGrath JJ, Burne THJ. 2017. Developmental vitamin D deficiency alters multiple neurotransmitter systems in the neonatal rat brain. International journal of developmental neuroscience : the official journal of the International Society for Developmental Neuroscience 62: 1-7 Khanna JM, Israel Y. 1980. Ethanol metabolism. International review of physiology 21: 275-315

164

Kim YS, Leventhal BL, Koh YJ, Fombonne E, Laska E, et al. 2011. Prevalence of autism spectrum disorders in a total population sample. The American journal of psychiatry 168: 904-12 Kishimoto R, Fujiwara I, Kitayama S, Goda K, Nakata Y. 1995. Changes in hepatic enzyme activities related to ethanol metabolism in mice following chronic ethanol administration. Journal of nutritional science and vitaminology 41: 527-43 Kleiber ML, Mantha K, Stringer RL, Singh SM. 2013. Neurodevelopmental alcohol exposure elicits long-term changes to gene expression that alter distinct molecular pathways dependent on timing of exposure. J Neurodev Disord 5: 6 Kleiber ML, Wright E, Singh SM. 2011. Maternal voluntary drinking in C57BL/6J mice: Advancing a model for fetal alcohol spectrum disorders. Behav Brain Res 223: 376- 87 Ko P, Burkert R, McGrath J, Eyles D. 2004. Maternal vitamin D3 deprivation and the regulation of apoptosis and cell cycle during rat brain development. Brain research. Developmental brain research 153: 61-8 Kolevzon A, Weiser M, Gross R, Lubin G, Knobler HY, et al. 2006. Effects of season of birth on autism spectrum disorders: Fact or fiction? The American journal of psychiatry 163: 1288-U3 Krueger DD, Howell JL, Hebert BF, Olausson P, Taylor JR, Nairn AC. 2006. Assessment of cognitive function in the heterozygous reeler mouse. Psychopharmacology (Berl) 189: 95-104 Kvaskoff D, Heath AK, Simila HA, Ko P, English DR, Eyles DW. 2016. Minimizing matrix effects for the accurate quantification of 25-hydroxyvitamin D metabolites in dried blood spots by LC-MS/MS. Clin Chem 62: 639 Kvaskoff D, Ko P, Simila HA, Eyles DW. 2012. Distribution of 25-hydroxyvitamin D3 in dried blood spots and implications for its quantitation by tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 901: 47 Laitinen K, Valimaki M. 1991. Alcohol and bone. Calcified tissue international 49 Suppl: S70-3 Landa RJ. 2008. Diagnosis of autism spectrum disorders in the first 3 years of life. Nat Clin Pract Neuro 4: 138-47 Lands WE. 1995. Alcohol and energy intake. Am J Clin Nutr 62: 1101s-06s Laroche F, Ramoz N, Leroy S, Fortin C, Rousselot-Paillet B, et al. 2008. Polymorphisms of coding trinucleotide repeats of homeogenes in neurodevelopmental psychiatric disorders. Psychiatr Genet 18: 295

165

Lehmann B, Meurer M. 2010. Vitamin D metabolism. Dermatol Ther 23: 2-12 Lesburguères E, Sparks FT, Reilly KC, Fenton AA. 2016. Active place avoidance is no more stressful than unreinforced exploration of a familiar environment. Hippocampus 26: 1481-85 Levey AI, Hersch SM, Rye DB, Sunahara RK, Niznik HB, et al. 1993. Localization of D1 and D2 dopamine receptors in brain with subtype-specific antibodies. Proc Natl Acad Sci U S A 90: 8861-5 Lévy F, Melo AI, Galef BG, Madden M, Fleming AS. 2003. Complete maternal deprivation affects social, but not spatial, learning in adult rats. Dev Psychobiol 43: 177-91 Li H, Yamagata T, Mori M, Momoi MY. 2005. Absence of causative mutations and presence of autism-related allele in FOXP2 in Japanese autistic patients. Brain & development 27: 207-10 Li T, Zeng Z, Zhao Q, Wang T, Huang K, et al. 2013. FoxP2 is significantly associated with schizophrenia and major depression in the Chinese Han population. World J Biol Psychiatry 14: 146-50 Li Y-X, Yang H-T, Marzena Z, Jason S, Yi Q, et al. 2007. Fetal alcohol exposure impairs hedgehog cholesterol modification and signaling. Lab Invest 87: 231-40 Lieber CS. 1991. Perspectives: do alcohol calories count? Am J Clin Nutr 54: 976-82 Light KE, Brown DP, Newton BW, Belcher SM, Kane CJM. 2002. Ethanol-induced alterations of neurotrophin receptor expression on Purkinje cells in the neonatal rat cerebellum. Brain Res 924: 71-81 Lindsley TA, Miller MW, Littner Y, Bearer CF. 2006. Signaling pathways regulating cell motility: a role in ethanol teratogenicity? Alcohol Clin Exp Res 30: 1445-50 Linnet KM, Dalsgaard S, Obel C, Wisborg K, Henriksen TB, et al. 2003. Maternal lifestyle factors in pregnancy risk of attention deficit hyperactivity disorder and associated behaviors: review of the current evidence. The American journal of psychiatry 160: 1028-40 Lisse TS, Liu T, Irmler M, Beckers J, Chen H, et al. 2011. Gene targeting by the vitamin D response element binding protein reveals a role for vitamin D in osteoblast mTOR signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 25: 937-47 Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, et al. 1997. Maternal care, hippocampal glucocorticoid receptors, and hypothalamic- pituitary-adrenal responses to stress. Science 277: 1659-62

166

Liu Y, Balaraman Y, Wang G, Nephew KP, Zhou FC. 2009. Alcohol exposure alters DNA methylation profiles in mouse embryos at early neurulation. Epigenet 4: 500-11 Livy DJ, Elberger AJ. 2008. Alcohol exposure during the first two trimesters-equivalent alters the development of corpus callosum projection neurons in the rat. Alcohol 42: 285-93 Livy DJ, Miller EK, Maier SE, West JR. 2003. Fetal alcohol exposure and temporal vulnerability: effects of binge-like alcohol exposure on the developing rat hippocampus. Neurotoxicol Teratol 25: 447-58 Lobellova V, Entlerova M, Svojanovska B, Hatalova H, Prokopova I, et al. 2013. Two learning tasks provide evidence for disrupted behavioural flexibility in an animal model of schizophrenia-like behaviour induced by acute MK-801: a dose-response study. Behav Brain Res 246: 55 Logge W, Kingham J, Karl T. 2014. Do individually ventilated cage systems generate a problem for genetic mouse model research? Genes, brain, and behavior 13: 713-20 Looker AC, Gunter EW. 1998. Hypovitaminosis D in medical inpatients. N Engl J Med 339: 344-5; author reply 45-6 Lord C, Risi S, Lambrecht L, Cook E, Leventhal B, et al. 2000. The autism diagnostic observation schedule—generic: A standard measure of social and communication deficits associated with the spectrum of autism. J Autism Dev Disord 30: 205-23 Love MI, Huber W, Anders S. 2014. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome biology 15: 550 Lugo JN, Marino MD, Cronise K, Kelly SJ. 2003. Effects of alcohol exposure during development on social behavior in rats. Physiol Behav 78: 185-94 Luo J, Miller MW. 1996. Ethanol inhibits basic fibroblast growth factor-mediated proliferation of c6 astrocytoma cells. J Neurochem 67: 1448-56 Lyall K, Schmidt RJ, Hertz-Picciotto I. 2014. Maternal lifestyle and environmental risk factors for autism spectrum disorders. Int J Epidemiol 43: 443-64 Maier SE, West JR. 2001. Regional differences in cell loss associated with binge-like alcohol exposure during the first two trimesters equivalent in the rat. Alcohol 23: 49- 57 Maldve RE, Zhang TA, Ferrani-Kile K, Schreiber SS, Lippmann MJ, et al. 2002. DARPP- 32 and regulation of the ethanol sensitivity of NMDA receptors in the nucleus accumbens. Nat Neurosci 5: 641-8

167

Mantha K, Kleiber ML, Singh SM. 2013. Neurodevelopmental timing of ethanol exposure may contribute to observed heterogeneity of behavioral deficits in a mouse model of Fetal Alcohol Spectrum Disorder (FASD). J Behav Brain Sci Vol.03No.01: 15 Marino MD, Aksenov MY, Kelly SJ. 2004. Vitamin E protects against alcohol-induced cell loss and oxidative stress in the neonatal rat hippocampus. International journal of developmental neuroscience : the official journal of the International Society for Developmental Neuroscience 22: 363-77 Marino MD, Cronise K, Lugo JN, Jr., Kelly SJ. 2002. Ultrasonic vocalizations and maternal-infant interactions in a rat model of fetal alcohol syndrome. Dev Psychobiol 41: 341-51 Marjonen H, Sierra A, Nyman A, Rogojin V, Grohn O, et al. 2015. Early maternal alcohol consumption alters hippocampal DNA methylation, gene expression and volume in a mouse model. PLoS One 10 Marks R. 1999. Sunscreens and vitamin D levels. J Am Acad Dermatol 40: 497-97 Marks R, McCarthy WH. 1990. Skin cancer: increasing incidence and public awareness. Med J Aust 153: 505 Marquardt K, Brigman JL. 2016. The impact of prenatal alcohol exposure on social, cognitive and affective behavioral domains: Insights from rodent models. Alcohol 51: 1-15 Marrone MC, Marinelli S, Biamonte F, Keller F, Sgobio CA, et al. 2006. Altered cortico- striatal synaptic plasticity and related behavioural impairments in reeler mice. Eur J Neurosci 24: 2061-70 Mason S, Anthony B, Lai X, Ringham HN, Wang M, et al. 2012. Ethanol exposure alters protein expression in a mouse model of Fetal Alcohol Spectrum Disorders. Int J Proteomics 2012: 867141 Matter K, Balda MS. 2003. Signalling to and from tight junctions. Nat Rev Mol Cell Biol 4: 225-37 Mattila P, Kiiskinen T, Lehikoinen K, Piironen V. 1999. Cholecalciferol and 25- hydroxycholecalciferol content of chicken egg yolk as affected by the cholecalciferol content of feed. J Agric Food Chem 47: 4089-92 Mattson SN, Crocker N, Nguyen TT. 2011. Fetal Alcohol Spectrum Disorders: Neuropsychological and behavioral features. Neuropsychol Rev 21: 81-101 Mattson SN, Goodman AM, Caine C, Delis DC, Riley EP. 1999. Executive functioning in children with heavy prenatal alcohol exposure. Alcohol Clin Exp Res 23: 1808

168

McCarthy-Jones S, Green MJ, Scott RJ, Tooney PA, Cairns MJ, et al. 2014. Preliminary evidence of an interaction between theFOXP2gene and childhood emotional abuse predicting likelihood of auditory verbal hallucinations in schizophrenia. J Psychiatr Res 50: 66-72 McClintick JN, Brooks AI, Deng L, Liang L, Wang JC, et al. 2014. Ethanol treatment of lymphoblastoid cell lines from alcoholics and non-alcoholics causes many subtle changes in gene expression. Alcohol 48: 603-10 McGivern RF, Raum WJ, Salido E, Redei E. 1988. Lack of prenatal testosterone surge in fetal rats exposed to alcohol: alterations in testicular morphology and physiology. Alcohol Clin Exp Res 12: 243-47 McGrath J, Saha S, Welham J, El Saadi O, MacCauley C, Chant D. 2004. A systematic review of the incidence of schizophrenia: the distribution of rates and the influence of sex, urbanicity, migrant status and methodology. Bmc Med 2 McGrath JJ, Burne TH, Feron F, Mackay-Sim A, Eyles DW. 2010a. Developmental vitamin D deficiency and risk of schizophrenia: A 10-Year update. Schizophr Bull 36: 1073- 78 McGrath JJ, Eyles DW, Pedersen CB, Anderson C, Ko P, et al. 2010b. Neonatal vitamin D status and risk of schizophrenia a population-based case-control study. Arch Gen Psychiatry 67: 889-94 Means LW, Medlin CW, Hughes VD, Gray SL. 1984. Hyperresponsiveness to methylphenidate in rats following prenatal ethanol exposure. Neurobehavioral toxicology and teratology 6: 187-92 Mercer KE, Wynne RA, Lazarenko OP, Lumpkin CK, Hogue WR, et al. 2012. Vitamin D supplementation protects against bone loss associated with chronic alcohol administration in female mice. The Journal of pharmacology and experimental therapeutics 343: 401-12 Merewood DA, Mehta CS, Chen FT, Bauchner FH, Holick FM. 2009. Association between Vitamin D deficiency and primary cesarean section. J Clin Endocrinol Metab 94: 940-45 Meyer LS, Riley EP. 1986. Social play in juvenile rats prenatally exposed to alcohol. Teratol 34: 1-7 Miller MW. 1986. Effects of alcohol on the generation and migration of cerebral cortical neurons. Science 233: 1308-11 Minasyan A, Keisala T, Zou J, Zhang Y, Toppila E, et al. 2009. Vestibular dysfunction in vitamin D receptor mutant mice. J Steroid Biochem Mol Biol 114: 161-66

169

Miranda RC, Pietrzykowski AZ, Tang Y, Sathyan P, Mayfield D, et al. 2010. MicroRNAs: master regulators of ethanol abuse and toxicity? Alcohol Clin Exp Res 34: 575-87 Moechars D, Weston MC, Leo S, Callaerts-Vegh Z, Goris I, et al. 2006. Vesicular glutamate transporter VGLUT2 expression levels control quantal size and neuropathic pain. J Neurosci 26: 12055-66 Molinoff PB, Axelrod J. 1971. Biochemistry of catecholamines. Annual review of biochemistry 40: 465-500 Mombereau C, Medvedeva V, Ghosh T, Hervé D, French C, et al. 2013. FOXP2 in the nucleus accumbens regulates reward signaling and social behavior. Moore CL, Wong L, Daum MC, Leclair OU. 1997. Mother-infant interactions in two strains of rats: implications for dissociating mechanism and function of a maternal pattern. Dev Psychobiol 30: 301-12 Moore DB, Madorsky I, Paiva M, Barrow Heaton M. 2004. Ethanol exposure alters neurotrophin receptor expression in the rat central nervous system: Effects of prenatal exposure. Journal of neurobiology 60: 101-13 Morgan MY, Levine JA. 1988. Alcohol and nutrition. The Proceedings of the Nutrition Society 47: 85-98 Mortensen PB, Pedersen CB, Westergaard T, Wohlfahrt J, Ewald H, et al. 1999. Effects of family history and place and season of birth on the risk of schizophrenia. N Engl J Med 340: 603-08 Mostofsky DI, Yehuda S, Rabinovitz S, Carasso R. 2000. The control of blepharospasm by essential fatty acids. Neuropsychobiol 41: 154-7 Motomura N, Shimizu K, Shimizu M, Aoki-Komori S, Taniguchi K, et al. 2002. A comparative study of isolation-induced ultrasonic vocalization in rodent pups. Exp Anim 51: 187-90 Mukherjee R, Layton M, Yacoub E, Turk J. 2011. Autism and autistic traits in people exposed to heavy prenatal alcohol: data from a clinical series of 21 individuals and nested case control study. Advances in Mental Health and Intellectual Disabilities 5: 42-49 Mulligan MK, Ponomarev I, Boehm SL, 2nd, Owen JA, Levin PS, et al. 2008. Alcohol trait and transcriptional genomic analysis of C57BL/6 substrains. Genes, brain, and behavior 7: 677-89 Muñoz-Villegas P, Rodríguez VM, Giordano M, Juárez J. 2017. Risk-taking, locomotor activity and dopamine levels in the nucleus accumbens and medial prefrontal cortex in male rats treated prenatally with alcohol. Pharmacol Biochem Behav 153: 88-96

170

Murugan M, Harward S, Scharff C, Mooney R. 2013. Diminished FoxP2 levels affect dopaminergic modulation of corticostriatal signaling important to song variability. Neuron 80: 1464-76 Naassila M, Daoust M. 2002. Effect of prenatal and postnatal ethanol exposure on the developmental profile of mRNAs encoding NMDA receptor subunits in rat hippocampus. J Neurochem 80: 850-60 Nagy LE, Lakshman MR, Casey CA, Bearer CF. 2002. Ethanol and membrane protein trafficking: diverse mechanisms of ethanol action. Alcohol Clin Exp Res 26: 287-93 Nanson JL, Hiscock M. 1990. Attention Deficits in Children Exposed to Alcohol Prenatally. Alcohol Clin Exp Res 14: 656-61 Narla A, Ebert BL. 2010. Ribosomopathies: human disorders of ribosome dysfunction. Blood 115: 3196-205 Naveilhan P, Neveu I, Wion D, Brachet P. 1996. 1,25-dihydroxyvitamin D-3, an inducer of glial cell line-derived neurotrophic factor. Neuroreport 7: 2171-75 Nazarian A, Rodarte-Freeman AL, McDougall SA. 1999. Dopaminergic modulation of kappa opioid-mediated ultrasonic vocalization, antinociception, and locomotor activity in the preweanling rat. Behav Neurosci 113: 816-25 Neckameyer WS, Weinstein JS. 2005. Stress affects dopaminergic signaling pathways in Drosophila melanogaster. Stress (Amsterdam, Netherlands) 8: 117-31 Nemere I, Garbi N, Hammerling G, Hintze KJ. 2012. Role of the 1,25D3-MARRS receptor in the 1,25(OH)2D3-stimulated uptake of calcium and phosphate in intestinal cells. Steroids 77: 897-902 Nesbit MA, Hannan FM, Howles SA, Babinsky VN, Head RA, et al. 2013. Mutations affecting G-protein subunit alpha11 in hypercalcemia and hypocalcemia. N Engl J Med 368: 2476-86 Nesby O, Dell S, Scanlon KS, Cogswell ME, Gillespie C, et al. 2002. Hypovitaminosis D prevalence and determinants among African American and white women of reproductive age: third National Health and Nutrition Examination Survey, 1988- 1994. Am J Clin Nutr 76: 187-92 Neveu I, Naveilhan P, Baudet C, Brachet P, Metsis M. 1994. 1,25-Dihydroxyvitamin D-3, regulates Nt-3, Nt-4 but not Bdnf messenger-Rna in astrocytes. Neuroreport 6: 124- 26 Newbury DF, Bonora E, Lamb JA, Fisher SE, Lai CSL, et al. 2002. FOXP2 is not a major susceptibility gene for autism or specific language impairment. Am J Med Genet 70: 1318-27

171

Ngo AS, M.; Issler, E., Thomas, J., Idrus, N. 2013. The effects of vitamin D on stress in rats exposed to alcohol during development. Presented at Society for Neuroscience, New Orleans, LA O'Tuathaigh CM, Babovic D, O'Sullivan GJ, Clifford JJ, Tighe O, et al. 2007. Phenotypic characterization of spatial cognition and social behavior in mice with 'knockout' of the schizophrenia risk gene neuregulin 1. Neurosci 147: 18-27 O'Tuathaigh CM, Harte M, O'Leary C, O'Sullivan GJ, Blau C, et al. 2010. Schizophrenia- related endophenotypes in heterozygous neuregulin-1 'knockout' mice. Eur J Neurosci 31: 349-58 O’Leary-Moore SK, Parnell SE, Lipinski RJ, Sulik KK. 2011. Magnetic resonance-based imaging in animal models of Fetal Alcohol Spectrum Disorder. Neuropsychol Rev 21: 167-85 Ogilvie KM, Rivier C. 1997. Prenatal alcohol exposure results in hyperactivity of the hypothalamic-pituitary-adrenal axis of the offspring: modulation by fostering at birth and postnatal handling. Alcohol Clin Exp Res 21: 424-9 Ostermeyer U, Schmidt T. 2006. Vitamin D and provitamin D in fish: determination by HPLC with electrochemical detection. Eur Food Res Technol 222: 403 Otis EM, Brent R. 1954. Equivalent ages in mouse and human embryos. Anat Rec 120: 33-63 Ouimet CC, Lamantia AS, Goldman-Rakic P, Rakic P, Greengard P. 1992. Immunocytochemical localization of DARPP-32, a dopamine and cyclic- AMP- regulated phosphoprotein, in the primate brain. J Comp Neurol 323: 209-18 Ouimet CC, Langley-Gullion KC, Greengard P. 1998. Quantitative immunocytochemistry of DARPP-32-expressing neurons in the rat caudatoputamen. Brain Res 808: 8-12 Overeem K, Eyles DW, McGrath JJ, Burne TH. 2016. The impact of vitamin D deficiency on behaviour and brain function in rodents. Curr Opin Behav Sci 7: 47-52 Padovani A, Cosseddu M, Premi E, Archetti S, Papetti A, et al. 2010. The speech and language FOXP2 gene modulates the phenotype of frontotemporal lobar degeneration. JAD 22: 923 Pappalardo-Carter DL, Balaraman S, Sathyan P, Carter ES, Chen W-JA, Miranda RC. 2013. Suppression and epigenetic regulation of miR-9 contributes to ethanol teratology: Evidence from zebrafish and murine fetal neural models. Alcohol Clin Exp Res 37: 1657-67

172

Parnell SE, Holloway HE, Baker LK, Styner MA, Sulik KK. 2014. Dysmorphogenic effects of first trimester‐equivalent ethanol exposure in mice: A magnetic resonance microscopy‐based study. Alcohol Clin Exp Res 38: 2008-14 Parnell SE, Oleary-Moore SK, Godin EA, Dehart DB, Johnson BW, et al. 2009. Magnetic resonance microscopy defines ethanol-induced brain abnormalities in prenatal mice: Effects of acute insult on gestational day 8. Alcohol Clin Exp Res 33: 1001-11 Perera F, Herbstman J. 2011. Prenatal environmental exposures, epigenetics, and disease. Reprod Toxicol 31: 363-73 Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. 2016. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protocols 11: 1650-67 Pietrzykowski AZ, Friesen RM, Martin GE, Puig SI, Nowak CL, et al. 2008. Posttranscriptional regulation of BK channel splice variant stability by miR-9 underlies neuroadaptation to alcohol. Neuron 59: 274-87 Pioggia G, Tonacci A, Tartarisco G, Billeci L, Muratori F, et al. 2014. Autism and lack of D3 vitamin: A systematic review. Res Autism Spectr Disord 8: 1685-98 Procopio M, Marriott PK. 1998. Seasonality of birth in epilepsy: a Danish study. Acta neurologica Scandinavica 98: 297-301 Procopio M, Marriott PK, Davies RJE. 2006. Seasonality of birth in Epilepsy: A Southern Hemisphere study. Seizure 15: 17-21 Procopio M, Marriott PK, Williams P. 1997. Season of birth: aetiological implications for epilepsy. Seizure 6: 99-105 Przybelski R, Agrawal S, Krueger D, Engelke J, Walbrun F, Binkley N. 2008. Rapid correction of low vitamin D status in nursing home residents. Osteoporos Int 19: 1621-28 Qi Y, Li X, Chang C, Xu F, He Q, et al. 2017. Ribosomal protein L23 negatively regulates cellular apoptosis via the RPL23/Miz-1/c- circuit in higher-risk myelodysplastic syndrome. Scientific reports 7: 2323 Raghuram S, Stayrook KR, Huang P, Rogers PM, Nosie AK, et al. 2007. Identification of heme as the ligand for the orphan nuclear receptors REV-ERBalpha and REV- ERBbeta. Nature structural & molecular biology 14: 1207-13 Ramanathan R, Wilkemeyer MF, Mittal B, Perides G, Charness ME. 1996. Alcohol inhibits cell-cell adhesion mediated by human L1 published erratum appears in J Cell Biol 1996 Jun;133(5):1139-40. J Cell Biol 133: 381-90

173

Rapin I, Tuchman RF. 2008. Autism: definition, neurobiology, screening, diagnosis. Pediatr Clin North Am 55: 1129-46 Rathbun W, Druse MJ. 1985. Dopamine, serotonin, and acid metabolites in brain regions from the developing offspring of ethanol‐treated rats. J Neurochem 44: 57-62 Ribases M, Sanchez-Mora C, Ramos-Quiroga J, Bosch R, Gomez N, et al. 2012. An association study of sequence variants in the forkhead box P2 (FOXP2) gene and adulthood attention-deficit/hyperactivity disorder in two European samples. Psychiatr Genet 22: 155-60 Ricceri L, Cutuli D, Venerosi A, Scattoni ML, Calamandrei G. 2007. Neonatal cholinergic hypofunction affects ultrasonic vocalizations and fear conditioning responses in preweaning rats. Behav Brain Res 183: 111-7 Rice D, Barone S, Jr. 2000. Critical periods of vulnerability for the developing nervous system: evidence from humans and animal models. Environ Health Perspect 108 Suppl 3: 511-33 Robinson M, Mattes E, Oddy WH, Pennell CE, van Eekelen A, et al. 2011. Prenatal stress and risk of behavioral morbidity from age 2 to 14 years: The influence of the number, type, and timing of stressful life events. Dev Psychopathol 23: 507-20 Robinson M, Oddy WH, McLean NJ, Jacoby P, Pennell CE, et al. 2010. Low–moderate prenatal alcohol exposure and risk to child behavioural development: a prospective cohort study. BJOG 117: 1139-52 Rojas-Mayorquín AE, Padilla-Velarde E, Ortuño-Sahagún D. 2016. Prenatal alcohol exposure in rodents as a promising model for the study of ADHD molecular basis. Front Neurosci 10 Ross AC. 2011. Vitamin D The 2011 report on dietary reference intakes for calcium and vitamin D. Public Health Nutr 14: 938-39 Ruegsegger U, Blank D, Keller W. 1998. Human pre-mRNA cleavage factor Im is related to spliceosomal SR proteins and can be reconstituted in vitro from recombinant subunits. Molecular cell 1: 243-53 Ryu HW, Park CW, Ryu KY. 2014. Disruption of polyubiquitin gene Ubb causes dysregulation of neural stem cell differentiation with premature gliogenesis. Scientific reports 4: 7026 Saad K, Abdel-Rahman AA, Elserogy YM, Al-Atram AA, Cannell JJ, et al. 2015. Vitamin D status in autism spectrum disorders and the efficacy of vitamin D supplementation in autistic children. Nutr Neurosci

174

Sakai Y, Shaw CA, Dawson BC, Dugas DV, Al-Mohtaseb Z, et al. 2011. Protein interactome reveals converging molecular pathways among autism disorders. Science translational medicine 3: 86ra49 Salaspuro MP, Lieber CS. 1978. Non-uniformity of blood ethanol elimination: its exaggeration after chronic consumption. Annals of clinical research 10: 294-7 Salinger WL, Ladrow P, Wheeler C. 2003. Behavioral phenotype of the reeler mutant mouse: effects of RELN gene dosage and social isolation. Behav Neurosci 117: 1257-75 Sampson HW. 1997. Alcohol, osteoporosis, and bone regulating hormones. Alcohol Clin Exp Res 21: 400-03 Sampson PD, Bookstein FL, Barr HM, Streissguth AP. 1994. Prenatal alcohol exposure, birthweight, and measures of child size from birth to age 14 years. Am J Public Health 84: 1421-28 Sanchez Vega MC, Chong S, Burne TH. 2013. Early gestational exposure to moderate concentrations of ethanol alters adult behaviour in C57BL/6J mice. Behav Brain Res 252: 326-33 Sanjuan J, Tolosa A, Gonzalez JC, Aguilar EJ, Perez-Tur J, et al. 2006. Association between FOXP2 polymorphisms and schizophrenia with auditory hallucinations. Psychiatr Genet 16: 67-72 Saporito MS, Brown ER, Hartpence KC, Wilcox HM, Vaught JL, Carswell S. 1994. Chronic 1,25-Dihydroxyvitamin D-3-mediated induction of nerve growth-factor messenger- RNA and protein in L929 fibroblasts and in adult-rat brain. Brain Res 633: 189-96 Sathyan P, Golden HB, Miranda RC. 2007. Competing interactions between micro-RNAs determine neural progenitor survival and proliferation after ethanol exposure: evidence from an ex vivo model of the fetal cerebral cortical neuroepithelium. J Neurosci 27: 8546 Savage DD, Becher M, de La Torre AJ, Sutherland RJ. 2002. Dose-dependent effects of prenatal ethanol exposure on synaptic plasticity and learning in mature offspring. Alcohol Clin Exp Res 26: 1752 Sayal K, Heron J, Golding J, Emond A. 2007. Prenatal alcohol exposure and gender differences in childhood mental health problems: A longitudinal population-based study. Pediatrics 119: e426-e34 Scattoni M, Ricceri L, Crawley J. 2011. Unusual repertoire of vocalizations in adult BTBR T plus tf/J mice during three types of social encounters. Genes Brain Behav. 10: 44- 56

175

Scattoni ML, Branchi I. 2009a. Vocal repertoire in mouse pups: strain differences, pp. 89- 95: Elsevier Science & Technology Scattoni ML, Crawley J, Ricceri L. 2009b. Ultrasonic vocalizations: a tool for behavioural phenotyping of mouse models of neurodevelopmental disorders. Neurosci Biobehav Rev 33: 508-15 Schaaf CP, Sabo A, Sakai Y, Crosby J, Muzny D, et al. 2011. Oligogenic heterozygosity in individuals with high-functioning autism spectrum disorders. Hum Mol Genet 20: 3366-75 Schmittgen TD, Livak KJ. 2008. Analyzing real-time PCR data by the comparative C-T method. Nat. Protocols 3: 1101-08 Schneider ML, Moore CF, Barnhart TE, Larson JA, DeJesus OT, et al. 2005. Moderate- level prenatal alcohol exposure alters striatal dopamine system function in rhesus monkeys. Alcohol Clin Exp Res 29: 1685-97 Schroeder BC, Hechenberger M, Weinreich F, Kubisch C, Jentsch TJ. 2000. KCNQ5, a Novel Potassium Channel Broadly Expressed in Brain, Mediates M-type Currents. The Journal of biological chemistry 275: 24089-95 Schuster I. 2011. Cytochromes P450 are essential players in the vitamin D signaling system. Biochim Biophys Acta 1814: 186-99 Scott L, Forssberg H, Aperia A, Diaz-Heijtz R. 2005. Locomotor effects of a D1R agonist are DARPP-32 dependent in adult but not weanling mice. Pediatr Res 58: 779-83 Sekar A, Bialas AR, de Rivera H, Davis A, Hammond TR, et al. 2016. Schizophrenia risk from complex variation of complement component 4. Nature 530: 177-83 Serrano M, Han M, Brinez P, Linask KK. 2010. Fetal alcohol syndrome: cardiac birth defects in mice and prevention with folate. Am J Clin Exp Obstet Gynecol 203: 75.e7 Shen R-Y, Hannigan J, Chiodo L. 1995. The effects of chronic amphetamine treatment on prenatal ethanol-induced changes in dopamine receptor function: Electrophysiological findings. The Journal of pharmacology and experimental therapeutics 274: 1054-60 Shen RY, Hannigan JH, Kapatos G. 1999. Prenatal ethanol reduces the activity of adult midbrain dopamine neurons. Alcohol Clin Exp Res 23: 1801-07 Shetty AK, Burrows RC, Phillips DE. 1993. Alterations in neuronal development in the substantia nigra pars compacta following in utero ethanol exposure: Immunohistochemical and Golgi studies. Neurosci 52: 311-22

176

Shu W, Cho JY, Jiang Y, Zhang M, Weisz D, et al. 2005. Altered ultrasonic vocalization in mice with a disruption in the Foxp2 gene. Proc Natl Acad Sci U S A 102: 9643-8 Siddiqui MF, Elwell C, Johnson MH. 2016. Mitochondrial dysfunction in Autism Spectrum Disorders. Autism Open access 6: 1000190 Siegel A, Malkowitz L, Moskovits MJ, Christakos S. 1984. Administration of 1,25- dihydroxyvitamin D3 results in the elevation of hippocampal seizure threshold levels in rats. Brain Res 298: 125-9 Silverman J, L. , Tolu S, S. , Barkan C, L. , Crawley J, N. . 2009. Repetitive self-grooming behavior in the BTBR mouse model of autism is blocked by the mGluR5 antagonist MPEP. Neuropsychopharmacol 35: 976 Singh SP, Snyder AK, Pullen GL. 1989. Maternal alcohol ingestion inhibits fetal glucose uptake and growth. Neurotoxicol Teratol 11: 215-9 Smiley JF, Saito M, Bleiwas C, Masiello K, Ardekani B, et al. 2015. Selective reduction of cerebral cortex GABA neurons in a late gestation model of fetal alcohol spectrum disorder. Alcohol (Fayetteville, N.Y.) 49: 571-80 Smith SM, Garic A, Berres ME, Flentke GR. 2014. Genomic factors that shape craniofacial outcome and neural crest vulnerability in FASD. Front Genet 5: 224 Soares AR, Pereira PM, Ferreira V, Reverendo M, Simoes J, et al. 2012. Ethanol exposure induces upregulation of specific microRNAs in zebrafish embryos. Toxicological sciences : an official journal of the Society of Toxicology 127: 18-28 Sokol RJ, Delaney-Black V, Nordstrom B. 2003. Fetal alcohol spectrum disorder. JAMA 290: 2996-99 Sood B, Delaney-Black V, Covington C, Nordstrom-Klee B, Ager J, et al. 2001. Prenatal alcohol exposure and childhood behavior at age 6 to 7 years: I. dose-response effect. Pediatrics 108: E34 Spanagel R, Weiss F. 1999. The dopamine hypothesis of reward: past and current status. Trends Neurosci 22: 521-27 Spaniel F, Horáček J, Tintěra J, Ibrahim I, Novák T, et al. 2011. Genetic variation in FOXP2 alters grey matter concentrations in schizophrenia patients. Neurosci Lett 493: 131-35 Spiteri E, Konopka G, Coppola G, Bomar J, Oldham M, et al. 2007. Identification of the transcriptional targets of FOXP2, a gene linked to speech and language, in developing human brain. Am J Med Genet 81: 1144-57 Squillace M, Dodero L, Federici M, Migliarini S, Errico F, et al. 2014. Dysfunctional dopaminergic neurotransmission in asocial BTBR mice. Transl Psychiatry 4: e427

177

Stanford SC. 2007. The open field test: reinventing the wheel. J Psychopharmacol 21: 134-35 Stipp CS, Litwack ED, Lander AD. 1994. Cerebroglycan: an integral membrane heparan sulfate proteoglycan that is unique to the developing nervous system and expressed specifically during neuronal differentiation. J Cell Biol 124: 149-60 Stoof JC, Kebabian JW. 1981. Opposing roles for D-1 and D-2 dopamine receptors in efflux of cyclic AMP from rat neostriatum. Nature 294: 366-68 Stuchlik A, Rezacova L, Vales K, Bubenikova V, Kubik S. 2004. Application of a novel Active Allothetic Place Avoidance task (AAPA) in testing a pharmacological model of psychosis in rats: comparison with the Morris Water Maze. Neurosci Lett 366: 162-66 Sugita S, Ho A, Sudhof TC. 2002. NECABs: a family of neuronal Ca(2+)-binding proteins with an unusual domain structure and a restricted expression pattern. Neurosci 112: 51-63 Suri D, Veenit V, Sarkar A, Thiagarajan D, Kumar A, et al. 2013. Early stress evokes age- dependent biphasic changes in hippocampal neurogenesis, BDNF expression, and cognition. Biol Psychiatry 73: 658-66 Suzuki M, Yoshioka M, Hashimoto M, Murakami M, Noya M, et al. 2013. Randomized, double-blind, placebo-controlled trial of vitamin D supplementation in Parkinson disease. Am J Clin Nutr 97: 1004 Svenningsson P, Nishi A, Fisone G, Girault J-A, Nairn AC, Greengard P. 2004. DARPP- 32: An integrator of neurotransmission. Annu Rev Pharmacol Toxicol 44: 269-96 Swanson DJ, King MA, Walker DW, Heaton MB. 1995. Chronic prenatal ethanol exposure alters the normal ontogeny of choline acetyltransferase activity in the rat septohippocampal system. Alcohol Clin Exp Res 19: 1252-60 Swartz ME, Wells MB, Griffin M, McCarthy N, Lovely CB, et al. 2014. A screen of zebrafish mutants identifies ethanol-sensitive genetic loci. Alcohol Clin Exp Res 38: 694-703 Tal TL, Franzosa JA, Tilton SC, Philbrick KA, Iwaniec UT, et al. 2012. MicroRNAs control neurobehavioral development and function in zebrafish. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 26: 1452-61 Tare M, Parkington H, Morley R. 2006. Vitamin D in pregnancy and offspring health In Early Life Origins of Health and Disease, ed. Wintour EM, Owens J, pp. 195-203: Springer US

178

Teramitsu I, White SA. 2006. FoxP2 regulation during undirected singing in adult songbirds. J Neurosci 26: 7390 Tesic D, Hawes JE, Zosky GR, Wyrwoll CS. 2015. Vitamin D deficiency in BALB/c mouse pregnancy increases placental transfer of glucocorticoids. Endocrinol 156: 3673-79 Theorell H, Mc KJ. 1961. Mechanism of action of liver alcohol dehydrogenase. Nature 192: 47-50 Thompson KN, Phillips LJ, Komesaroff P, Yuen HP, Wood SJ, et al. 2007. Stress and HPA-axis functioning in young people at ultra high risk for psychosis. J Psychiatr Res 41: 561-69 Toda Y, Mori K, Hashimoto T, Miyazaki M, Nozaki S, et al. 2006. Administration of secretin for autism alters dopamine metabolism in the central nervous system. Brain & development 28: 99-103 Tolosa A, Sanjuan J, Dagnall AM, Molto MD, Herrero N, de Frutos R. 2010. FOXP2 gene and language impairment in schizophrenia: association and epigenetic studies. BMC Med Genet 11: 114 Tong M, Ziplow J, Chen WC, Nguyen Q-G, Kim CM, de la Monte SM. 2013. Motor function deficits following chronic prenatal ethanol exposure are linked to impairments in insulin/igf, notch and wnt signaling in the cerebellum. Diabetes Metab J 4: 238 Tonkiss J, Bonnie KE, Hudson JL, Shultz PL, Duran P, Galler JR. 2003. Ultrasonic call characteristics of rat pups are altered following prenatal malnutrition. Dev Psychobiol 43: 90-101 Toso L, Roberson R, Woodard J, Abebe D, Spong CY. 2006. Prenatal alcohol exposure alters GABA Aα 5 expression: A mechanism of alcohol-induced learning dysfunction. Am J Clin Exp Obstet Gynecol 195: 522-27 Tran TD, Cronise K, Marino MD, Jenkins WJ, Kelly SJ. 2000. Critical periods for the effects of alcohol exposure on brain weight, body weight, activity and investigation. Behav Brain Res 116: 99-110 Trinko JRL, B. B.; Solecki, W. B.; Wickham, R. J.; Tellez, L. A.; De Araujo, I. E.; Addy, N. A.; Dileone, R. J. 2013. A role for vitamin D in diet-induced obesity and dopamine- related behavior. Presented at Society for Neuroscience, New Orleans, LA Tsui D, Vessey JP, Tomita H, Kaplan DR, Miller FD. 2013. FoxP2 regulates neurogenesis during embryonic cortical development. J Neurosci 33: 244 Turner CA, Calvo N, Frost DO, Akil H, Watson SJ. 2008. The fibroblast growth factor system is downregulated following social defeat. Neurosci Lett 430: 147-50

179

Uban KA, Comeau WL, Ellis LA, Galea LA, Weinberg J. 2013. Basal regulation of HPA and dopamine systems is altered differentially in males and females by prenatal alcohol exposure and chronic variable stress. Psychoneuroendocrino 38: 1953-66 Ulug S, Riley EP. 1983. The effect of methylphenidate on overactivity in rats prenatally exposed to alcohol. Neurobehavioral toxicology and teratology 5: 35-9 Valenzuela CF, Puglia MP, Zucca S. 2011. Focus on: neurotransmitter systems. Alcohol Res Health 34: 106 Vallés S, Felipo V, Montoliu C, Guerri C. 1995. Alcohol exposure during brain development reduces 3H-MK-801 binding and enhances metabotropic-glutamate receptor- stimulated phosphoinositide hydrolysis in rat hippocampus. Life Sci 56: 1373-83 Van Cromphaut SJ, Dewerchin M, Hoenderop JG, Stockmans I, Van Herck E, et al. 2001. Duodenal calcium absorption in vitamin D receptor-knockout mice: functional and molecular aspects. Proc Natl Acad Sci U S A 98: 13324-9 van den Buuse M. 2009. Modeling the positive symptoms of schizophrenia in genetically modified mice: Pharmacology and methodology aspects. Schizophr Bull 36: 246-70 Vangipuram SD, Lyman WD. 2012. Ethanol affects differentiation‐related pathways and suppresses wnt signaling protein expression in human neural stem cells. Alcohol Clin Exp Res 36: 788-97 Vernes SC, Oliver P, Spiteri E, Lockstone H, Puliyadi R, et al. 2011. Foxp2 regulates gene networks implicated in neurite outgrowth in the developing brain. Plos Genet Vernes SC, Spiteri E, Nicod J, Groszer M, Taylor JM, et al. 2007. High-throughput analysis of promoter occupancy reveals direct neural targets of FOXP2, a gene mutated in speech and language disorders. Am J Med Genet 81: 1232-50 Vilensky JA, Damasio AR, Maurer RG. 1981. Gait disturbances in patients with autistic behavior: a preliminary study. Arch Neurol 38: 646-9 Vinkhuyzen AAE, Eyles DW, Burne THJ, Blanken LME, Kruithof CJ, et al. 2017. Gestational vitamin D deficiency and autism spectrum disorder. BJPsych Open 3: 85-90 Wainwright P, Gagnon M. 1985. Moderate prenatal ethanol exposure interacts with strain in affecting brain development in BALBc and C57BL6 mice. Exp Neurol 88: 84-94 Walaas SI, Aswad DW, Greengard P. 1983. A dopamine- and cyclic AMP-regulated phosphoprotein enriched in dopamine-innervated brain regions. Nature 301: 69-71 Walaas SI, Greengard P. 1984. DARPP-32, a dopamine- and adenosine 3':5'- monophosphate-regulated phosphoprotein enriched in dopamine-innervated brain regions. I. Regional and cellular distribution in the rat brain. J Neurosci 4: 84-98

180

Wallen-Mackenzie A, Nordenankar K, Fejgin K, Lagerstrom MC, Emilsson L, et al. 2009. Restricted cortical and amygdaloid removal of vesicular glutamate transporter 2 in preadolescent mice impacts dopaminergic activity and neuronal circuitry of higher brain function. J Neurosci 29: 2238-51 Walsh RN, Cummins RA. 1976. The open field test: a critical review. Psychological bulletin 83: 482-504 Wang G, Bieberich E. 2010. Prenatal alcohol exposure triggers ceramide-induced apoptosis in neural crest-derived tissues concurrent with defective cranial development. Cell Death Dis 1: e46 Wang J-J, Li Y. 2016a. KCNQ potassium channels in sensory system and neural circuits. Acta Pharmacol Sin 37: 25-33 Wang L, Wu L, Wang X, Deng J, Ma Z, et al. 2015. Prenatal alcohol exposure inducing the apoptosis of mossy cells in hippocampus of SMS2−/− mice. Environ Toxicol Pharmacol 40: 975-82 Wang LL, Zhang Z, Li Q, Yang R, Pei X, et al. 2009. Ethanol exposure induces differential microRNA and target gene expression and teratogenic effects which can be suppressed by folic acid supplementation. Human reproduction (Oxford, England) 24: 562-79 Wang T, Shan L, Du L, Feng J, Xu Z, et al. 2016b. Serum concentration of 25- hydroxyvitamin D in autism spectrum disorder: a systematic review and meta- analysis. Eur Child Adolesc Psychiatry 25: 341-50 Wang X, Mozhui K, Li Z, Mulligan MK, Ingels JF, et al. 2012. A promoter polymorphism in the Per3 gene is associated with alcohol and stress response. Transl Psychiatry 2: e73 Ward OB, Ward IL, Denning JH, Hendricks SE, French JA. 2002. Hormonal mechanisms underlying aberrant sexual differentiation in male rats prenatally exposed to alcohol, stress, or both. Arch Sex Behav 31: 9-16 Warren RP, Burger RA, Odell D, Torres AR, Warren WL. 1994. Decreased plasma concentrations of the C4B complement protein in autism. Archives of pediatrics & adolescent medicine 148: 180-3 Warren RP, Odell JD, Warren WL, Burger RA, Maciulis A, Torres AR. 1995. Is decreased blood plasma concentration of the complement C4B protein associated with attention-deficit hyperactivity disorder? J Am Acad Child Adolesc Psychiatry 34: 1009-14

181

Webb AR, Kline L, Holick MF. 1988. Influence of season and latitude on the cutaneous synthesis of vitamin D3: exposure to winter sunlight in Boston and Edmonton will not promote vitamin D3 synthesis in human skin. J Clin Endocrinol Metab 67: 373- 78 Webster WS, Walsh DA, McEwen SE, Lipson AH. 1983. Some teratogenic properties of ethanol and acetaldehyde in C57BL/6J mice: Implications for the study of the fetal alcohol syndrome. Teratol 27: 231-43 Wellmann KA, George F, Brnouti F, Mooney SM. 2015. Docosahexaenoic acid partially ameliorates deficits in social behavior and ultrasonic vocalizations caused by prenatal ethanol exposure. Behav Brain Res 286: 201-11 Wesierska M, Dockery C, Fenton AA. 2005. Beyond memory, navigation, and inhibition: behavioral evidence for hippocampus-dependent cognitive coordination in the rat. J Neurosci 25: 2413-9 Whitehouse AJO, Holt BJ, Serralha M, Holt PG, Hart PH, Kusel MMH. 2013. Maternal vitamin D levels and the autism phenotype among offspring. J Autism Dev Disord 43: 1495-504 Wilcke A, Ligges C, Burkhardt J, Alexander M, Wolf C, et al. 2011. Imaging genetics of FOXP2 in dyslexia. Eur J Hum Gen 20: 224 Wilkins C, Sheline Y, Roe CM, Birge SJ, Morris J. 2006. Vitamin D deficiency is associated with low mood, worse cognitive performance and smaller hippocampal volumes in older adults. Neuropsychopharmacol 31: S133-S33 Willoughby KA, Sheard ED, Nash K, Rovet J. 2008. Effects of prenatal alcohol exposure on hippocampal volume, verbal learning, and verbal and spatial recall in late childhood. J Int Neuropsychol Soc 14: 1022-33 Wilson SE, Cudd TA. 2011. Focus on: The use of animal models for the study of Fetal Alcohol Spectrum Disorders. Alcohol Res Health 34: 92-98 Winslow JT, Insel TR. 1990. Serotonergic modulation of rat pup ultrasonic vocal development: studies with 3,4-methylenedioxymethamphetamine. The Journal of pharmacology and experimental therapeutics 254: 212-20 Wion D, MacGrogan D, Neveu I, Jehan F, Houlgatte R, Brachet P. 1991. 1,25- Dihydroxyvitamin D3 is a potent inducer of nerve growth factor synthesis. J Neurosci Res 28: 110-4 Wojcik SM, Rhee JS, Herzog E, Sigler A, Jahn R, et al. 2004. An essential role for vesicular glutamate transporter 1 (VGLUT1) in postnatal development and control of quantal size. Proc Natl Acad Sci U S A 101: 7158-63

182

Wozniak DF, Hartman RE, Boyle MP, Vogt SK, Brooks AR, et al. 2004. Apoptotic neuro degeneration induced by ethanol in neonatal mice is associated with profound learning/memory deficits in juveniles followed by progressive functional recovery in adults. Neurobiol Dis 17: 403-14 Wu J, Zou H, Strong J, Yu J, Zhou X, et al. 2005. Bimodal effects of MK-801 on locomotion and stereotypy in C57BL/6 mice. Psychopharmacology 177: 256-63 Wu WQ, Beilhartz G, Roy Y, Richard CL, Curtin M, et al. 2010. Nuclear translocation of the 1,25D(3)-MARRS (membrane associated rapid response to steroids) receptor protein and NF kappa B in differentiating NB4 leukemia cells. Exp Cell Res 316: 1101-08 Yamada L. 2017. A study of the consequences of early gestational ethanol and/or nicotine exposure in the mouse. The University of Queensland, School of Medicine Yamanaka Y, Kitano A, Takao K, Prasansuklab A, Mushiroda T, et al. 2011. Inactivation of fibroblast growth factor binding protein 3 causes anxiety-related behaviors. Molecular and cellular neurosciences 46: 200-12 Yang M, Abrams DN, Zhang JY, Weber MD, Katz AM, et al. 2012. Low sociability in BTBR T+tf/J mice is independent of partner strain. Physiol Behav 107: 649-62 Yin L, Wu N, Lazar MA. 2010. Nuclear receptor Rev-erbalpha: a heme receptor that coordinates circadian rhythm and metabolism. Nuclear receptor signaling 8: e001 Yoshida T, Ohki S, Kanazawa M, Mizunuma H, Kikuchi Y, et al. 1998. Inhibitory effects of prostaglandin D2 against the proliferation of human colon cancer cell lines and hepatic metastasis from colorectal cancer. Surgery today 28: 740-5 Young J, Kavanagh M, Anderson G, Shaywitz B, Cohen D. 1982. Clinical neurochemistry of autism and associated disorders. J Autism Dev Disord 12: 147-65 Yus-nájera E, Muñoz A, Salvador N, Jensen BS, Rasmussen HB, et al. 2003. Localization of KCNQ5 in the normal and epileptic human temporal neocortex and hippocampal formation. Neurosci 120: 353-64 Zakhari S. 2006. Overview: how is alcohol metabolized by the body? Alcohol Res Health 29: 245-54 Zanatta L, Goulart PB, Goncalves R, Pierozan P, Winkelmann-Duarte EC, et al. 2012. 1alpha,25-dihydroxyvitamin D(3) mechanism of action: modulation of L-type calcium channels leading to calcium uptake and intermediate filament phosphorylation in cerebral cortex of young rats. Biochim Biophys Acta 1823: 1708-19 Zerbo O, Iosif A-M, Delwiche L, Walker C, Hertz-Picciotto I. 2011. Month of Conception and Risk of Autism. Epidemiol 22: 469-75

183

Zhang CR. 2013. Brain structure and function in a mouse model of moderate prenatal ethanol exposure. The University of Queensland, School of Medicine U6 - Dissertation Zhang CR, Ho MF, Vega MC, Burne TH, Chong S. 2015. Prenatal ethanol exposure alters adult hippocampal VGLUT2 expression with concomitant changes in promoter DNA methylation, H3K4 trimethylation and miR-467b-5p levels. Epigenetics & chromatin 8: 40 Zhou FC, Sari Y, Powrozek T, Goodlett CR, Li T-K. 2003. Moderate alcohol exposure compromises neural tube midline development in prenatal brain. Dev Brain Res 144: 43-55 Zhou FC, Zhao Q, Liu Y, Goodlett CR, Liang T, et al. 2011. Alteration of gene expression by alcohol exposure at early neurulation. BMC genomics 12: 124 Zhu Y, Zhou R, Yang R, Zhang Z, Bai Y, et al. 2012. Abnormal neurogenesis in the dentate gyrus of adult mice lacking 1,25-dihydroxy vitamin D3 (1,25-(OH)2 D3). Hippocampus 22: 421-33 Zimmerberg B, Rosenthal AJ, Stark AC. 2003. Neonatal social isolation alters both maternal and pup behaviors in rats. Dev Psychobiol 42: 52-63 Zink M, Arac G, Frank ST, Gass P, Gebicke-Haater PJ, Spanagel R. 2009. Perinatal exposure to alcohol reduces the expression of complexins I and II. Neurotoxicol Teratol 31: 400-05 Zink M, Ferbert T, Frank ST, Seufert P, Gebicke-Haerter PJ, Spanagel R. 2011. Perinatal exposure to alcohol disturbs spatial learning and glutamate transmission-related gene expression in the adult hippocampus. Eur J Neurosci 34: 457-68

184

Appendices

Appendix 1. List of primers List of oligonucleotides used in this thesis. Primers were obtained from previous literature, or pre-designed and purchased from Sigma Aldrich, Australia.

Primer name Forward Sequence Reverse Sequence Aadc CCTTGTTGCAGATAGGACCG GAAGCCCTGGAGAGACAA Cntnap2 TGGGGAAGACCAGAGATGAG TCTGAGAGCAGGCTACAGGG D2R CTGGAAGCCTCAAGCAGC ATTCAGTGGATCCATCAGGG Darpp-32 AACGCCGAGTTCTGGGGATA CGGCCAATGAGCATCGAGAA Disc1 CACAGTGTTTGCCTTCATGC CAGAAGGCTAGGCATCCTCA Drd1 TTTCTGGTGCCCAAGACAGT AGAAGTCCCTCTCCACCACC FoxP2 TTGGCTGCTTAAAGTGCTCAT GGACTCCACTCCAAGGTTGA Grin3b ATGGTCTCCAGGGGACTAGC GTGCTGAGCGTCCTACGG Hprt1 CAGTACAGCCCCAAAATGGT TTGCGCTCATCTTAGGCTTT Itga3 TGAGGGGACACAGGTACACA AGACTGAGCGACAACAGCG Itgb1 CAATCCAATCCAGGAAACCA ACACCGACCCGAGACCCT Itgb3 GGCGTTGTTGTTGGAGAGTC GCCTCACTGACTGGGAACTC Maoa GGGGGCTGTCATCAAGTGCAT TGGCAGGCATTGACCCATCTG Mef2c GGATGAGCGTAACAGACAGGT ATCAGTGCAATCTCACAGTCG Reln GAGAGCTGACCACCACATGC GCTCACATTGGAAGGGGCTC Tbr1 GGTCAAGCGGTCCATGTC CTGTCACCGCCTACCAGAAC Th CGCCGTCCAATGAACCTT CACTATGCCCACCCCCAG Vglut1 TGTTCCTCATAGCCTCCC GTCCTCCATTTCACTTTCGT Vglut2 CCCTGGAGGTGCCTGAGAA GCGGTGGATAGTGCTGTTGTT Vglut3 CACATCCTTGCCTGTCTAT GACCCACCTTACTTATTGC Vmat2 ACGTGCAAGTTGGTCTGTTG CGCAAATATGGGAATTGGAT

185

Appendix 2. Group x Sex interactions in novelty-induced locomotion There were no significant interactions of Group x Sex in adult novelty-induced locomotion.

Locomotion P70 F Sig. (p) Sex 1.42 0.24 Sex x Diet 0.45 0.50 Sex x Exposure 0.36 0.55 Strain x Sex x Diet 1.44 0.23 Strain x Sex x Exposure 0.47 0.50 Sex x Diet x Exposure 0.17 0.68 Strain x Sex x Diet x Exposure 3.27 0.07

186

Appendix 3. Group x Sex interactions in adult hippocampal gene expression There were no significant interactions of Group x Sex in Vglut1-3 adult hippocampal expression.

P70 qPCR F Sig. (p) Sex VGLUT1 2.08 0.15 VGLUT2 0.61 0.44 VGLUT3 1.66 0.20 Strain x Sex VGLUT1 0.28 0.60 VGLUT2 1.67 0.20 VGLUT3 0.81 0.37 Diet x Sex VGLUT1 0.74 0.39 VGLUT2 0.37 0.54 VGLUT3 0.03 0.86 Exposure x Sex VGLUT1 0.36 0.55 VGLUT2 0.09 0.77 VGLUT3 0.45 0.50 Strain x Diet x Sex VGLUT1 1.64 0.21 VGLUT2 0.03 0.86 VGLUT3 0.35 0.55 Strain x Exposure x Sex VGLUT1 0.24 0.62 VGLUT2 1.45 0.23 VGLUT3 0.01 0.91 Diet x Exposure x Sex VGLUT1 0.02 0.88 VGLUT2 0.87 0.35 VGLUT3 0.31 0.58 Strain x Diet x Exposure x Sex VGLUT1 0.01 0.93 VGLUT2 0.13 0.72 VGLUT3 1.41 0.24

187

Appendix 4. Top molecular and cellular functions affected by DVD deficiency and PEE on gene expression

Molecular and cellular functions Std-H2O v. DVD-H2O Name p-value #Molecules Molecular Transport 4.95E-02 - 2.50E-06 17 Small Molecule Biochemistry 4.95E-02 - 2.50E-06 16 Amino Acid Metabolism 1.83E-02 - 3.12E-05 5 Drug Metabolism 2.06E-02 - 3.09E-04 7 Cell Death and Survival 4.96E-02 - 1.33E-03 10

Std-H2O v. Std-PEE Amino Acid Metabolism 1.33E-02 - 9.41E-04 1 Molecular Transport 1.33E-02 - 9.41E-04 2 Small Molecule Biochemistry 1.33E-02 - 9.41E-04 3 Cell-To-Cell Signalling and Interaction 3.60E-02 - 1.88E-03 1 Cell Death and Survival 6.81E-03 - 2.12E-03 1

Std-H2O v. DVD-PEE Cell Signalling 1.39E-02 - 6.43E-05 3 Nucleic Acid Metabolism 2.31E-02 - 6.43E-05 4 Small Molecule Biochemistry 3.22E-02 - 6.43E-05 15 Cellular Assembly and Organization 2.76E-02 - 4.84E-04 23 Cellular Function and Maintenance 3.22E-02 - 4.87E-04 21

188

Appendix 5. Top transcripts up and downregulated by DVD deficiency and PEE

Top Analysis-Ready Molecules Log Ratio up-regulated Log Ratio down-regulated Molecules Expr. Value Molecules Expr. Value Std-H2O v. DVD-H2O KCNQ5 0.355 PTGDS -0.335 NECAB1 0.334 CRABP2 -0.332 HEY2 0.333 CDH1 -0.281 HYDIN 0.325 PPP1R13L -0.276 WDR78 0.323 CD3EAP -0.274 Slco1a4 0.321 Actn3 -0.271 P2RY12 0.321 ALDOB -0.26 CRYM 0.315 MAGEL2 -0.254 PVT1 0.311 ADAT3 -0.253 THRB 0.309 SHPK -0.252

Std-H2O v. Std-PEE C4A/C4B* 0.497 FGFBP3 -0.292 Ubb 0.454 RPL23 -0.211 Gm6682 0.412 CRYM 0.312

Std-H2O v. DVD-PEE NR1D1 0.436 F13A1 -0.518 PER3 0.404 Pisd-ps1 -0.469 NR1D2 0.376 Pisd-ps2 -0.465 ARSG 0.352 Tubb2a-ps2 -0.436 SCUBE2 0.336 Gm6654 -0.431 NEUROD1 0.325 DENND1C -0.425 THBS2 0.32 OSR1 -0.419 RNPC3 0.315 LAMC3 -0.4 Rps15a-ps4 0.315 COL6A3 -0.392 Gm14057 0.311 C16orf89 -0.382

189

Appendix 6. Top molecular and cellular functions altered by DVD deficiency and PEE on protein expression

Molecular and cellular functions Std-H2O v. DVD-H2O Name p-value #Molecules RNA Post-Transcriptional Modification 2.01E-02 - 1.02E-05 9 Cell Death and Survival 3.41E-02 - 1.22E-04 21 Protein Synthesis 3.13E-02 - 2.67E-04 17 Cellular Function and Maintenance 3.39E-02 - 4.44E-04 17 Molecular Transport 3.12E-02 - 6.65E-04 15

Std-H2O v. Std-PEE Cell Morphology 1.94E-02 - 1.42E-06 15 Cellular Assembly and Organization 1.94E-02 - 1.42E-06 18 Cellular Development 1.73E-02 - 1.42E-06 10 Cellular Function and Maintenance 2.16E-02 - 1.42E-06 16 Cellular Growth and Proliferation 1.73E-02 - 1.42E-06 10

Std-H2O v. DVD-PEE RNA Post-Transcriptional Modification 1.83E-02 - 5.32E-05 8 Carbohydrate Metabolism 3.63E-02 - 2.17E-04 9 Protein Synthesis 4.34E-02 - 5.35E-04 13 Cell Morphology 4.87E-02 - 7.39E-04 13 Cell Death and Survival 4.87E-02 - 9.71E-04 10

190

Appendix 7. Top proteins up and downregulated by DVD deficiency and PEE

Top Analysis-Ready Molecules Log Ratio up-regulated Log Ratio down-regulated Molecules Expr. Value Molecules Expr. Value Std-H2O v. DVD-H2O ETFA 4.355 GNA11 -2.139 COTL1 3.234 RPL7 -1.221 NUDT21 2.887 SKP1 -1.221 VAPA 1.765 TNIK -1.199 NUP85 1.443 VPS35 -1.025 CORO1C 1.307 ATP1B1 -0.99 LARP1 1.231 OSGEP -0.954 DGKE 1.078 UFM1 -0.95 MTPN 1.076 FSD1 -0.937 CMAS 1.063 UBFD1 -0.86

Std-H2O v. Std-PEE NUDT21 1.978 GPC2 -2.374 SCAMP1 1.813 MARS -1.968 DEK 1.547 SKP1 -1.598 Gm9755 1.161 DNM3 -1.055 SURF4 1.048 FAF1 -1.029 LRRC40 1.038 SRCIN1 -0.897 COTL1 1.03 TRA2A -0.797 COPS4 1.014 FSD1 -0.734 SMU1 1.014 CDC23 -0.596 ACO1 0.849 APOE -0.584

Std-H2O v. DVD-PEE LARP1 2.586 PSMD13 -1.944 COTL1 2.085 PSMA5 -1.391 FLNA 1.88 ETFA -1.353 SURF4 1.506 RPL7 -1.058 LRRC40 1.404 SRGAP3 -0.985 SLC25A46 1.173 FKBP3 -0.943 NUDT21 1.166 Ndufs5 -0.886 SEPT7 0.92 UFM1 -0.748 ADD1 0.869 RPS6 -0.707 ACO1 0.845 IGSF5 -0.693

191