The Effects of Enriched Environment on Paraventricular Hypothalamic Brain-derived. Neurotrophic Factor Expression

Similar documents
BIOMEDICAL SCIENCES GRADUATE PROGRAM SPRING 2017

Role of the ventromedial hypothalamic Steroidogenic Factor 1/ Adrenal 4. glucose metabolism in mice.

Insulin-Leptin Interactions

BIOMEDICAL SCIENCES GRADUATE PROGRAM SPRING 2017

Bi156 lecture 2, 1/6/12. Eating and weight regulation

CNS Control of Food Intake. Adena Zadourian & Andrea Shelton

Hypothalamus. Small, central, & essential.

Method of leptin dosing, strain, and group housing influence leptin sensitivity in high-fat-fed weanling mice

Table 1. Oligonucleotides and RT-PCR conditions Supplementary Material and Methods Fig. 1

Internal Regulation II Energy

Figure 1: The leptin/melanocortin pathway Neuronal populations propagate the signaling of various molecules (leptin, insulin, ghrelin) to control

Copyright 2017 The Guilford Press

Protocol: specimen preparation (brain dissection and region extraction) for using RT-qPCR to examine gene expression in brain regions of a fish

Chronic Stimulation of Leptin on Food Intake and Body Weight after Microinjection into the Ventromedial Hypothalamus of Conscious Rats

Follow this and additional works at: Part of the Psychology Commons

Introduction. Leptin secretion after a high-fat meal in normal-weight rats: strong predictor of long-term body fat accrual on a high-fat diet

Sex Differences in Central Expression and Effects of Brain-Derived Neurotrophic Factor

Ingestive Behaviors 33. Introduction. Page 1. control and story lines. (a review of general endocrinology) Integration (or the basic reflex arc model)

Homeostasis and Mechanisms of Weight Regulation

Hypothalamic TLR2 triggers sickness behavior via a microglia-neuronal axis

Investigation of the role of nesfatin-1/nucb2 in the central nervous system. Ph.D. thesis Katalin Könczöl

Neurophysiology of the Regulation of Food Intake and the Common Reward Pathways of Obesity and Addiction. Laura Gunter

Ghrelin mediates stressinduced. behavior in mice. Chuang et al 2011 L3: Love, Lust, Labor

Neural pathways controlling homeostatic and hedonic feeding in rats on free-choice diets van den Heuvel, J.K.

Motivation 1 of 6. during the prandial state when the blood is filled

COMPENSATION OF MELANOCORTIN-3 AND MELANOCORTIN-5 RECEPTORS IN THE BRAIN OF MELANOCORTIN-4 RECEPTOR KNOCKOUT RATS. A thesis submitted to the

Peripubertal, leptin-deficient ob/ob female mice were used in an investigation of

Microarray profiling of gene expression after sleep deprivation and recovery sleep 03/05/2008

SORCS1 and SORCS3 control energy balance and orexigenic peptide production

Early Life Effects on Hypothalamic Circuitry. By: The Mystery Gang

SUPPLEMENTARY INFORMATION

FLASH CARDS. Kalat s Book Chapter 10 Alphabetical

Nature Neuroscience: doi: /nn Supplementary Figure 1. Diverse anorexigenic signals induce c-fos expression in CEl PKC-δ + neurons

LESSON 3.3 WORKBOOK. How do we decide when and how much to eat?

Chapter 12. Ingestive Behavior

Supplementary Information Titles Journal: Nature Medicine

Hypothalamus. To learn how the brain regulates neuroendocrine secretions NTA Ch 14, pgs Key Figs: 14-3; 14-4,

Taylor Yohe. Project Advisor: Dr. Martha A. Belury. Department of Human Nutrition at the Ohio State University

Browning of white adipose tissue: role of hypothalamic signaling

Supplemental methods. Total RNA was extracted from the stomach, liver, pancreas, pituitary, and

Ingestive Behavior: Feeding & Weight Regulation. Hypovolemic vs. Osmotic Thirst

Metabolic Programming. Mary ET Boyle, Ph. D. Department of Cognitive Science UCSD

SUPPLEMENTARY INFORMATION

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

YK052 Mouse Leptin ELISA

Supplementary Methods

Temperature, Regulation, Thirst, and Hunger

Standard Operating Procedure

Rat Leptin ELISA FOR LABORATORY USE ONLY YANAIHARA INSTITUTE INC AWAKURA, FUJINOMIYA - SHI SHIZUOKA, JAPAN

Table S1. qpcr primer list.

Supplemental data Supplemental Figure Legends Supplemental Figure 1. Supplemental Figure 2.

Hormonal Regulation of Food Intake

Rat Leptin-HS ELISA FOR LABORATORY USE ONLY YANAIHARA INSTITUTE INC AWAKURA, FUJINOMIYA - SHI SHIZUOKA, JAPAN

Satiety induced by neuropeptide FF and gastrin in birds. Amanda Lynn Logan

Chapter 1. General introduction. Part of this chapter is adapted from Adan et.al., Br.J.Pharmacol. 2006;149:815

Ingestive Behaviors 21. Introduction. Page 1. control and story lines. (a review of general endocrinology) Integration (or the basic reflex arc model)

Zhu et al, page 1. Supplementary Figures

Ophthalmology, Radiation Oncology,

Environmental and Genetic Activation of a Brain-Adipocyte BDNF/Leptin Axis Causes Cancer Remission and Inhibition

a Supplementary Figure 1 Celastrol Withaferin A Individual drugs

Induction of brain-region specific forms of obesity by Agouti

1 Neuroregulation of Appetite

Environmental and Genetic Activation of a Brain-Adipocyte BDNF/Leptin Axis Causes Cancer Remission and Inhibition

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity

Νευροφυσιολογία και Αισθήσεις

Prasad S. Dalvi. Copyright by Prasad S. Dalvi 2012

ARTICLE A Self-Study Tutorial using the Allen Brain Explorer and Brain Atlas to Teach Concepts of Mammalian Neuroanatomy and Brain Function

Supplementary Figure 1

A Tryptophan Hydroxylase Inhibitor Decreases Hepatic FGF21 Expression and Circulating FGF21 in Mice Fed A High-Fat Diet

Mice lacking the syndecan-3 gene are resistant to diet-induced obesity

Management of Obesity. Objectives. Background Impact and scope of Obesity. Control of Energy Homeostasis Methods of treatment Medications.

Stressin with Ghrelin. Peptide Pods: Lara, Elijah, Karen, Brandon, Milena, Max & Lucile

letters to nature ... AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus

Supplemental Information Supplementary Table 1. Tph1+/+ Tph1 / Analyte Supplementary Table 2. Tissue Vehicle LP value

Figure S1. Body composition, energy homeostasis and substrate utilization in LRH-1 hep+/+ (white bars) and LRH-1 hep-/- (black bars) mice.

Central nervous system control of food intake

Supplementary Figure 1. DJ-1 modulates ROS concentration in mouse skeletal muscle.

Ambient Temperature Stabilization of RNA derived from Jurkat, HeLa and HUVEC Cell Lines for Use in RT-qPCR Assays

Behavioral and Motivational mechanisms of Brain. Limbic system and the Hypothalamus

Nature Neuroscience: doi: /nn Supplementary Figure 1. Visualization of AT1a-positive cells using AT1a lacz/+ mouse.

Epithelial interleukin-25 is a key mediator in Th2-high, corticosteroid-responsive

Activity Patterns in the Rat Olfactory Bulb in Response to Straight-Chain Aliphatic Odorants Differing in Carbon Number

THE ROLE OF INSULIN RECEPTOR SIGNALING IN THE BRAIN. COGS 163 By: Pranav Singh Alexandra Villar

Chapter 24 Cholesterol, Energy Balance and Body Temperature. 10/28/13 MDufilho

SUPPLEMENTARY INFORMATION

BMI risk SNPs associate with increased CADM1 and CADM2 expression in the cerebellum of human subjects.

Central injection of fibroblast growth factor 1 induces sustained remission of diabetic hyperglycemia in rodents

GPR120 *** * * Liver BAT iwat ewat mwat Ileum Colon. UCP1 mrna ***

HEK293FT cells were transiently transfected with reporters, N3-ICD construct and

Energy Balance. Applied Human Metabolism VII. Energy Out. Factors that effect BMR/RMR 17/03/2016

Comp Question 2012 NRSA proposal

RNA extraction, RT-PCR and real-time PCR. Total RNA were extracted using

The levels of mrna expression in the mouse brain were measured at 52 dpi after the brains were divided into five regions: cortex, hippocampus,

I. Introduction. II. Characteristics

Fat-Pad Specific Effects of Lipectomy on Appetitive and Consummatory Ingestive Behaviors in Siberian Hamsters (Phodopus sungorus)

Nature Neuroscience: doi: /nn Supplementary Figure 1. Splenic atrophy and leucopenia caused by T3 SCI.

Protocols. Harvesting and Sectioning the Ascending Aorta

Gene expression of NPY and POMC in the arcuate nucleus of adult male rats subjected to early postnatal food restriction

Distinct effects of leptin and melanocortin agonist in medial. hypothalamic nuclei on glucose uptake in peripheral tissues

Examination of Thyrotropin-Releasing Hormone (TRH)-synthesizing System in the Rodent Hypothalamus

Transcription:

The Effects of Enriched Environment on Paraventricular Hypothalamic Brain-derived Neurotrophic Factor Expression An Undergraduate Honors Research Thesis Presented in partial fulfillment of the requirements for graduation with Honors Research Distinction in Neuroscience in the undergraduate colleges of The Ohio State University By Xinyue Yao The Ohio State University July 2016 Project Advisor: Dr. Lei Cao, Dept. of Molecular Virology, Immunology & Medical Genetics

ABSTRACT The regulation of energy homeostasis involves a balance between food intake and energy expenditure. The hypothalamus is a crucial regulator for energy balance and consists of several discrete nuclei, including the arcuate (ARC), ventromedial (VMH), dorsomedial (DMH), and paraventricular (PVH) hypothalamus. Brain-derived neurotrophic factor (BDNF) expressed in hypothalamic nuclei is significant in regulating energy homeostasis. Our studies demonstrated that an enriched environment (EE), a complex housing with social and physical stimulation, upregulated BDNF expression in the ARC and VMH/DMH leading to an anti-obesity and anti-tumor phenotype. Moreover, the EE anti-obesity phenotype can be mimicked by overexpression of ARC and VMH/DMH Bdnf. While studies have shown diet- and geneticinduced obesity can be reversed by transferring the Bdnf gene to ARC and VMH/DMH, the roles of other nuclei in mediating the EE phenotype is not known. Research has shown that PVH BDNF has a strong and lasting impact on suppressing food intake and increasing energy expenditure, and the deletion of Bdnf in the PVH leads to hyperphagia and severe obesity. Thus, we hypothesized PVH Bdnf is upregulated by EE. Our results did not demonstrate significant upregulation of Bdnf expression in PVH or ARC after EE exposure. However, we observed a markedly increase of Vgf (non-acronymic) expression in PVH of EE mice, suggesting Vgf can be our next target gene of interest to study EE s anti-obesity effects. INTRODUCTION Obesity is one of the leading health problems in the U.S., with a prevalence of more than 78.6 million adults and more than one-third of children and adolescents (Ogden, Carroll, Kit, & Flegal, 2014; Ogden et al., 2016). Obesity is associated with increased risk of cardiovascular diseases, depression and cancers (Skinner, Perrin, Moss, & Skelton, 2015; Wong, Janssen, & Ross, 2003). The economic burden of obesity is greater than one-fifth of all annual medical expenses, with a $14.1 billion obese-related medical expenditure simply on children and 2

adolescents (Hammond & Levine, 2010; Trasande & Chatterjee, 2009). Therefore, finding effective treatments for obesity is in urgent need. Obesity is generally resulted from excess energy intake or few energy expenditures. The energy homeostasis is controlled by genetic and environmental factors. One of the genetic components that control energy balance is the hypothalamus. The hypothalamus is a crucial regulator for energy balance. Several discrete nuclei in the hypothalamus are involved in energy balance control circuits: arcuate nucleus (ARC), ventromedial hypothalamus (VMH), dorsomedial hypothalamus (DMH), and paraventricular hypothalamus (PVH). Moreover, many studies found that brain-derived neurotrophic factor (BDNF), a widely-expressed member of the neurotrophin family related to growth factors in the brain, is highly involved in hypothalamic energy control circuits. The deletion of Bdnf and its receptor tyrosine kinase receptor B (trkb/ntrk2) in the hypothalamus can induce severe obesity phenotypes (Gray et al., 2006). Studies also demonstrated that Bdnf knockout in hypothalamic nuclei can induce obesity phenotypes, whereas Bdnf overexpression can rescue the mutant obese phenotype (Abizaid, Gao, & Horvath, 2006; Liao et al., 2012; Xu et al., 2003). Nevertheless, our group is approaching this association between BDNF and hypothalamic energy homeostasis circuits from the environmental aspect. Our study showed that an enriched environment (EE), -a complex housing supplemented with physical, social, and cognitive stimuli, induced resistance to diet-induced obesity and improved metabolism in obese mice (Cao et al., 2011). EE upregulates Bdnf expression in ARC, DMH, and VMH; overexpression of Bdnf in these nuclei mimics the EE anti-obesity and anti-cancer phenotype (Cao et al., 2009; 2010; Xiao et al., 2016). However, Bdnf expression in other hypothalamic nuclei and their association to EE effects are poorly investigated. A recent study showed Bdnf deletion in PVH induced hyperphagia and resulted in severe obesity (An, Liao, Kinney, Sahibzada, & Xu, 2015), suggesting PVH can be our candidate for obesity therapeutic target in EE. Thus, in this study we hypothesized that EE upregulates Bdnf expression in PVH. 3

METHODS Environmental Enrichment. Male 3-week-old C57BL/6 mice (from Charles River) were randomly assigned to control housing (HC) and enriched environment (EE) (n=10 per group). Ten mice were housed in two medium enriched bins of 73 cm 41 cm 46 cm (n=5 per bin). Enriched bins were supplemented with running wheels, tunnels, igloos, huts, and nesting material in addition to standard lab chow and water (Andrew M Slater, 2015) (Figure 1). Control mice were housed under standard laboratory conditions with five mice per cage. Food and water were changed weekly and provided to all mice ad libitum. Housing Figure 1. Top-down view of EE cages and static wires in both control and enrichment groups were cleaned every week. Rearrangement of enrichment devices were conducted weekly and all toys were cleaned each week. Measurement of body weight and food intake was recorded every week. All mice were housed in their respective environments for four weeks before being sacrificed. Cryosectioning. Mice were perfused with 4% paraformaldehyde (Sigma). Fixed brains were kept in phosphate-buffered saline (PBS) overnight at 4 ºC and were subsequently switched to PBS with 30% sucrose. Brains were placed in whole brain cryomolds filled with optimal cutting temperature (OCT) compound. Cryomolds were placed in dry ice until frozen, at which point brains were removed from the mold and sectioned on a ThermoFisher cryostat at -20 C into 15 μm slices. Sections were collected on slides and stored at -80 C until staining. 4

Histology and Microscopy. Slides were dried at room temperature for two hours before staining. The section was treated with xylene for 5 min, dehydrated with 95% ethanol, then 70%, washed with deionized water, treated with cresyl violet solution for 10 min (at 60 C), washed with distilled water, rehydrated with 70% ethanol, 95%, then 100%, treated with xylene for 5 min, and coverslipped using cytoseal. A 10x imaging of the section was captured on a Zeiss Axio Imager M1 microscope. Tissue Collection. Brain and adipose tissue were collected four weeks post housing. PVH and ARC nuclei were microdissected immediately upon isolation from skulls. The following adipose tissue depots were collected and weighed: brown adipose tissue (BAT), epididymal adipose tissue (EAT), inguinal adipose tissue (IAT), and retroperitoneal adipose tissue (RAT). Hypothalamic Microdissection. Brains were quickly isolated on ice. The slices containing PVH and ARC were sectioned from three 1-mm-thick coronal sections (-0.58 to -3.58 mm from bregma) with a mouse brain matrix. PVH and ARC were both microdissected with a 30 microsurgical knife (OASIS) under a dissection scope. PVH was dissected from an inverted triangular section 0.75 mm downwards from the ventral tip of the third ventricle and 0.75 mm bilateral from midline in the first slice. ARC was dissected from a rectangular section on the second and third slices. The height of the rectangular section was 0.50 mm upwards from the dorsal edge of hypothalamus, whereas the width was 0.75 mm and 0.50 mm bilateral from midline respectively. In the second slice, it contained a small portion of VMH, but this would be addressed as ARC as the majority was from ARC nucleus. Tissues were snap-frozen on dry ice and stored at -80 C until further analysis. 5

RNA Isolation and cdna Synthesis. QIAGEN RNeasy Micro Kit was used to extract RNA from PVH and ARC of selected HC and EE mice (n=5 per group). All procedures were conducted according to the manufacturer s protocol. The extracted RNA was eluted with 28 μl RNase-free water to attain a 26 μl RNA sample. A spectrophotometer (NanoDrop1000) was used to determine RNA quantity and quality. All RNA samples were used with 0.1 μg for cdna synthesis with TaqMan Reverse Transcription Reagent (Applied Biosystems). Real-time Quantitative Polymerase Chain Reaction (qpcr). Real-time qpcr was carried out using StepOnePlus System (Applied Biosystem) with the Power SYBR Green PCR Master Mix (Applied Biosystems). Primers were designed to detect the following genes in the PVH cdna: Bdnf, Crh, Crhr1, cfos, Ghrh, Mc4r, Ntrk2, Oxt, Sst, Th, Trh, Vgf. The following genes were detected in the ARC cdna: AgRP, Bdnf, Npy, Ntrk2, Vgf. Sequences of primers are available upon requests. Data were calibrated to endogenous control Hprt1 and the relative gene expression was quantified using the 2 - CT ΔΔ method. Statistical Analysis. Data were expressed as means ± SEM. We used Student s t test (two tails, unpaired) to analyze the food intake, body weight, adiposity and gene expression in HC and EE group, as well as in the relative gene expression in PVH and ARC, with a p-value < 0.05 considered statistically significant. 6

RESULTS PVH and ARC Microdissection. Our previous study used laser capture microdissection (LCM) for hypothalamic nuclei dissection (Cao et al., 2010; 2011). However, LCM required highly-skilled operators and is expensive in training and operating. For this study, we used microsurgical knife to dissect PVH and ARC. Firstly, we tested our accuracy of dissecting PVH and ARC with a mouse brain matrix and a microsurgical knife. We conducted Nissl-stained histology to learn the location of PVH in the hypothalamus on a separate group of 8- or 9-week-old spare mice. All mice brain histology sections were collected using a cryostat. The histology showed a dense nuclei population located bilaterally adjacent to the ventral tip of the third ventricle which is one of the landmark for PVH (Figure 2A), suggesting our result is consistent with the staining presented in the mice stereotaxic atlas. Thus, we were confident of dissecting the correct slice containing PVH. Secondly, to test the accuracy of our microdissection of PVH and ARC with stab knife, A B C 14 25 PVH ARC/VMH Relative mrna Levels 12 10 8 6 4 2 20 15 10 5 * ** 0 Crh Trh 0 AgRP Npy Figure 2. Mice Brain Section Histology and Gene Expression Levels of PVH and ARC in Spare Mice. (A) Representative Nissl-stained histology of mice brain section. (B) Relative CRH and TRH expression levels in PVH and ARC (n=3). (C) Relative AGRP and NPY expression levels in PVH and ARC (n=3; *p < 0.05; **p <0.01) Values are mean ± SEM. we ran real-time qpcr to examine the relative gene expressions in these two regions. Two of the major neuropeptidergic populations in PVH are corticotrophin-releasing hormone (CRH) and 7

thyrotropin-releasing hormone (TRH) (Biag et al., 2011). CRH and TRH were used as gene markers for PVH, and we expected higher Crh and Trh expressions in PVH than in ARC. The gene markers we used for ARC were neuropeptide Y (NPY) and agouti-gene related protein (AGRP). NPY mrna has the highest expression in ARC and Agrp is almost exclusively expressed in ARC (Chronwall, Chase, & O'Donohue, 1984; Parker & Bloom, 2012). Our results showed that expression of Agrp and Npy were drastically higher in ARC than in PVH (Figure 2B; *p <0.05; **p<0.01). The Crh and Trh expressions in PVH were approximately 6-fold higher than in ARC (Figure 2C). The qpcr results indicated that using microdissection stab knife could give us enough accuracy for our further analysis, and therefore we could start our enrichment Weekly Food Intake (g) 150 100 50 0 A HC EE * * 1 2 3 4 Time (week) Average Daily Food Intake (per animal/g/d) 5 4 3 2 1 0 B HC * EE Average Body Weight (g) 22 20 18 16 14 12 10 8 6 C HC EE 0 1 2 3 4 Time (week) Average Total Weight Gain (g) D 14.0 12.0 10.0 8.0 6.0 4.0 2.0 0.0 HC EE Figure 3. EE Increased Food Intakes Without Changing the Body Weight. (A). EE increases food intake at 3 weeks and 4 weeks (*p value< 0.05). (B). Average food intake of per animal per day. (* p value < 0.05). (C). Average body weight showed no change in EE. (D) The average of the total weight gains in HC and EE mice. Values are mean ± SEM. n=10 per group. 8

experiment confidently. EE Increases Food Intake without Changing the Body Weight. Twenty 3-week-old male C57BL/6 mice were randomized into control housing cages (HC) or mid-size enriched bins (EE) (n=5 per cage) for four weeks. Standard laboratory chow and water were provided to all mice ad libitum. Food intake was recorded from four cages weekly. At 3 weeks and 4 weeks, EE groups showed significantly increased food consumption compared to HC groups (Figure 3A; p <0.05). The average daily food intake for each EE mice was higher compared to their HC littermates (Figure 3B; p <0.05). Nevertheless, EE mice did not show difference in either weekly body weight or total weight gain than HC group (Figure 3C & 3D). The results suggested that EE induced more energy expenditure but not suppressed food intake. EE Decreases Adiposity in EE Mice. All mice were sacrificed after four-week s exposure to their respective housing environment. The adipose and brain tissues of the mice were collected. We measured the weight of the adipose tissue, and calibrated fat pad mass to body weight. Our results demonstrated that EE decreased adiposity, with a drastic reduction in BAT Fat Pad Weigh / Body Weight 0.014 0.012 0.01 0.008 0.006 0.004 0.002 * HC EE ** ** (p<0.05), EAT and RAT (p<0.01) (Figure 4). Thus, we can confirm that EE reduced adiposity even 0 BAT IAT EAT RAT Figure 4. EE Decreases Adiposity in Mice. The average of fat pad weight calibrated to body weight. *p < 0.05; **p < 0.01 n=10 per group 9

with an increasing food intake. This result is consistent with our previous study, in which largesize enriched bins were used (Cao et al., 2011), suggesting a mid-size enrichment bin can induce EE phenotype. ARC and PVH Gene Expression Profile In our previous studies, EE induced upregulation of Bdnf expression in ARC and VMH/DMH (Cao et al., 2010; 2011). In this study, we examined the relative expression of Bdnf and its receptor Ntrk2 together with genes expressed in ARC and PVH. One of the gene we examined in both ARC and PVH was Vgf, a nerve growth factor-inducible transcript. Our recent study suggested that VGF was acting downstream of hypothalamic BDNF and was involved in 3 Relative mrna Levels 2.5 2 1.5 1 ** * HC EE 0.5 0 AgRP Bdnf Npy Ntrk2 Vgf Figure 5. Gene Expression Profile in ARC. Values are mean ± SEM. *p <0.05; **p <0.07 (n=5 per group). the melanocortin central pathway (Foglesong et al., 2016). The melanocortin pathway integrates central hormonal and neuronal signals to control energy balance (Balthasar et al., 2005; Garfield, Lam, Marston, Przydzial, & Heisler, 2009). One of the melanocortin receptor, melanocortin receptor 4 (Mc4r), is highly expressed in PVH; PVH receives direct melanocortin 10

input and is downstream of ARC nucleus (Sutton, Myers, & Olson, 2016). Thus, Vgf expression in ARC and PVH may reveal some unknown association along the melanocortin pathway. AGRP and NPY were used as gene markers for ARC, both of which are neuropeptides involved in promoting feeding behavior (Krashes et al., 2014; Morton, Meek, & Schwartz, 2014). In ARC gene profile, an upregulation of Npy (p <0.05) and a trend of increasing Agrp (p <0.07) were observed (Figure 5), which was consistent with the observed increasing food intake in EE mice. Nevertheless, the upregulation of Bdnf expression in ARC was not observed, and no change was found in Vgf expression either (Figure 5). We examined the expression of Bdnf and its receptor Ntrk2 with some genes of interests in PVH, including Crh and one of its receptor Chrh1, c-fos, Trh, growth hormone-releasing hormone (Ghrh), Mc4r, oxytocin (Oxt), somatostatin (Sst), tyrosine hydroxylase (Th) (An et al., 2015; Bali & Kovacs, 2003). Our qpcr showed no change of PVH Bdnf expression in EE mice. 3 Relative mrna Levels 2.5 2 1.5 1 0.5 HC EE * 0 Bdnf Crh cfos Crhr1 Ghrh Mc4R Ntrk2 Oxt Sst Th Trh Vgf Figure 6. Gene Expression Profile in PVH. Values are mean ± SEM. *p <0.01. For Bdnf, Crh, Th, Vgf, n=4 (HC), n=5 (EE) 11

Among the genes examined, only Vgf showed significant upregulation in PVH of EE mice, suggesting PVH VGF might play a role in EE-associated metabolic change. DISCUSSION Our results demonstrate that modified enriched environment in mid-size bin markedly decreased adiposity even with increased food intake, and induced upregulation in Npy and Agrp expression after 4-weeks of EE exposure. This result suggests our mid-size enrichment induced similar EE effects to what we observed in large-size bin with a larger group of mice in our previous study (Cao et al., 2010). However, EE-induced upregulation of Bdnf expression in ARC was not observed in this study. One possible explanation is that the mid-size EE setting induced ARC Bdnf upregulation at an earlier time point, but this upregulation was not sustained at 4 weeks. Our previous study showed that EE upregulated ARC Bdnf at 2 weeks and sustained at 4 weeks, but the upregulation was not significant at 9 weeks (Cao et al., 2010), suggesting Bdnf upregulation was an early event responding to EE. Our animal group-size is smaller than the one in the previous study, which could lead to a weaker EE effect. Moreover, compared to our previous study used laser capture microdissection to dissect ARC nuclei, the limited accuracy of the microsurgical knife may also lead to an insignificant result. The EE effect on PVH gene expression was limited based on our study, and our hypothesized EE-induced PVH Bdnf regulation was not observed. One recent study demonstrates that Bdnf deletion in anterior PVH leads to hyperphagia, whereas the medial and posterior PVH BDNF is more involved in thermogenesis than food intake (An et al., 2015). Our previous data showed EE led to a slight but significant increase in the body temperature in dietinduced obese mouse model as well as an elevation of adipose thermogenesis (Cao et al., 2011). Therefore, EE might upregulate Bdnf in medial/posterior PVH but not in the anterior part. Our dissection was not accurate enough to separate the anterior and posterior PVH, and thus the possible EE-induced upregulation of Bdnf in medial/posterior PVH was not observed. 12

Moreover, our PVH gene profile showed an upregulation of VGF in EE mice. VGF is thought to be downstream of BDNF, which suggests PVH Bdnf could be upregulated at an earlier time point. Our results showed reduced adiposity and upregulated Npy and Agrp expression in ARC, suggesting an EE anti-obesity effect. In order to test whether PVH and ARC Bdnf upregulation occurs at an earlier time point in the mid-size enrichment, we can design an experiment to investigate EE effects at two week s point in mid-size bin. Moreover, we can employ laser caption microdissection to collect anterior and medial/posterior PVH to explore if the Bdnf expression levels in the two regions are differentially regulated in EE. Finally, we can examine VGF s sufficiency and necessity in EE effects, and thus to evaluate the potentiality of VGF as a therapeutic gene for obesity. ACKNOWLEDGEMENTS I would like to express my gratitude to Dr. Lei Cao for providing me a great learning opportunity. I would like to thank Jason Siu in helping me oversee the project, from laboratory training to data collection to editing my thesis. I thank Nicholas Queen and Run Xiao for assisting in tissue collection and in reviewing my thesis. I would like to acknowledge Grant Foglesong, Wei Huang, Xianglan Liu, Travis Mcmurphy, Andrew Slater, and Kyle Widstrom for laboratory training and technical assistance. Last but not least, I thank Seemaab Ali, Amanda Huang, and Fangli Zhao for editorial suggestions. 13

REFERENCES: Abizaid, A., Gao, Q., & Horvath, T. L. (2006). Thoughts for Food: Brain Mechanisms and Peripheral Energy Balance. Neuron, 51(6), 691 702. http://doi.org/10.1016/j.neuron.2006.08.025 An, J. J., Liao, G.-Y., Kinney, C. E., Sahibzada, N., & Xu, B. (2015). Discrete BDNF Neurons in the Paraventricular Hypothalamus Control Feeding and Energy Expenditure. Cell Metabolism, 22(1), 175 188. http://doi.org/10.1016/j.cmet.2015.05.008 Andrew M Slater, L. C. (2015). A Protocol for Housing Mice in an Enriched Environment. Journal of Visualized Experiments: JoVE, (100), e52874. http://doi.org/10.3791/52874 Bali, B., & Kovacs, K. J. (2003). GABAergic control of neuropeptide gene expression in parvocellular neurons of the hypothalamic paraventricular nucleus. European Journal of Neuroscience, 18(6), 1518 1526. http://doi.org/10.1046/j.1460-9568.2003.02877.x Balthasar, N., Dalgaard, L. T., Lee, C. E., Yu, J., Funahashi, H., Williams, T., et al. (2005). Divergence of Melanocortin Pathways in the Control of Food Intake and Energy Expenditure. Cell, 123(3), 493 505. http://doi.org/10.1016/j.cell.2005.08.035 Biag, J., Huang, Y., Gou, L., Hintiryan, H., Askarinam, A., Hahn, J. D., et al. (2011). Cyto- and chemoarchitecture of the hypothalamic paraventricular nucleus in the C57BL/6J male mouse: A study of immunostaining and multiple fluorescent tract tracing. The Journal of Comparative Neurology, 520(1), 6 33. http://doi.org/10.1002/cne.22698 Cao, L., Choi, E. Y., Liu, X., Martin, A., Wang, C., Xu, X., & During, M. J. (2011). White to Brown Fat Phenotypic Switch Induced by Genetic and Environmental Activation of a Hypothalamic- Adipocyte Axis. Cell Metabolism, 14(3), 324 338. http://doi.org/10.1016/j.cmet.2011.06.020 Cao, L., Lin, E.-J. D., Cahill, M. C., Wang, C., Liu, X., & During, M. J. (2009). Molecular therapy of obesity and diabetes by a physiological autoregulatory approach. Nature Medicine, 15(4), 447 454. http://doi.org/10.1038/nm.1933 Cao, L., Liu, X., Lin, E.-J. D., Wang, C., Choi, E. Y., Riban, V., et al. (2010). Environmental and Genetic Activation of a Brain-Adipocyte BDNF/Leptin Axis Causes Cancer Remission and Inhibition. Cell, 142(1), 52 64. http://doi.org/10.1016/j.cell.2010.05.029 Chronwall, B. M., Chase, T. N., & O'Donohue, T. L. (1984). Coexistence of neuropeptide Y and somatostatin in rat and human cortical and rat hypothalamic neurons. Neuroscience Letters, 52(3), 213 217. http://doi.org/10.1016/0304-3940(84)90164-2 Foglesong, G. D., Huang, W., Liu, X., Slater, A. M., Siu, J., Yildiz, V., et al. (2016). Role of Hypothalamic VGF in Energy Balance and Metabolic Adaption to Environmental Enrichment in Mice. Endocrinology, 157(3), 983 996. http://doi.org/10.1210/en.2015-1627 Garfield, A. S., Lam, D. D., Marston, O. J., Przydzial, M. J., & Heisler, L. K. (2009). Role of central melanocortin pathways in energy homeostasis. Trends in Endocrinology & Metabolism, 20(5), 203 215. http://doi.org/10.1016/j.tem.2009.02.002 Gray, J., Yeo, G. S. H., Cox, J. J., Morton, J., Adlam, A. L. R., Keogh, J. M., et al. (2006). Hyperphagia, Severe Obesity, Impaired Cognitive Function, and Hyperactivity Associated With Functional Loss of One Copy of the Brain-Derived Neurotrophic Factor (BDNF) Gene. Diabetes, 55(12), 3366 3371. http://doi.org/10.2337/db06-0550 Hammond, R., & Levine. (2010). The economic impact of obesity in the United States. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy, Volume 3, 285 11. http://doi.org/10.2147/dmsott.s7384 Krashes, M. J., Shah, B. P., Madara, J. C., Olson, D. P., Strochlic, D. E., Garfield, A. S., et al. (2014). An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature, 507(7491), 238 242. http://doi.org/10.1038/nature12956 Liao, G.-Y., An, J. J., Gharami, K., Waterhouse, E. G., Vanevski, F., Jones, K. R., & Xu, B. (2012). Dendritically targeted Bdnf mrna is essential for energy balance and response to leptin. Nature Medicine, 18(4), 564 571. http://doi.org/10.1038/nm.2687 14

Morton, G. J., Meek, T. H., & Schwartz, M. W. (2014). Neurobiology of food intake in health and disease. Nature Publishing Group, 15(6), 367 378. http://doi.org/10.1038/nrn3745 Ogden, C. L., Carroll, M. D., Kit, B. K., & Flegal, K. M. (2014). Prevalence of Childhood and Adult Obesity in the United States, 2011-2012. Jama, 311(8), 806 9. http://doi.org/10.1001/jama.2014.732 Ogden, C. L., Carroll, M. D., Lawman, H. G., Fryar, C. D., Kruszon-Moran, D., Kit, B. K., & Flegal, K. M. (2016). Trends in Obesity Prevalence Among Children and Adolescents in the United States, 1988-1994 Through 2013-2014. Jama, 315(21), 2292 8. http://doi.org/10.1001/jama.2016.6361 Parker, J. A., & Bloom, S. R. (2012). Hypothalamic neuropeptides and the regulation of appetite. Neuropharmacology, 63(1), 18 30. http://doi.org/10.1016/j.neuropharm.2012.02.004 Skinner, A. C., Perrin, E. M., Moss, L. A., & Skelton, J. A. (2015). Cardiometabolic Risks and Severity of Obesity in Children and Young Adults. Dx.Doi.org. http://doi.org/10.1056/nejmoa1502821 Sutton, A. K., Myers, M. G., Jr, & Olson, D. P. (2016). The Role of PVH Circuits in Leptin Action and Energy Balance. Annual Review of Physiology, 78(1), 207 221. http://doi.org/10.1146/annurev-physiol-021115-105347 Trasande, L., & Chatterjee, S. (2009). The Impact of Obesity on Health Service Utilization and Costs in Childhood. Obesity, 17(9), 1749 1754. http://doi.org/10.1038/oby.2009.67 Wong, S. L., Janssen, I., & Ross, R. (2003). Abdominal Adipose Tissue Distribution and Metabolic Risk. Sports Medicine, 33(10), 709 726. http://doi.org/10.2165/00007256-200333100-00001 Xiao, R., Bergin, S. M., Huang, W., Slater, A. M., Liu, X., Judd, R. T., et al. (2016). Environmental and Genetic Activation of Hypothalamic BDNF Modulates T-cell Immunity to Exert an Anticancer Phenotype. Cancer Immunology Research, 4(6), 488 497. http://doi.org/10.1158/2326-6066.cir-15-0297 Xu, B., Goulding, E. H., Zang, K., Cepoi, D., Cone, R. D., Jones, K. R., et al. (2003). Brainderived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nature Neuroscience, 6(7), 736 742. http://doi.org/10.1038/nn1073 15