Manipulation of Corticotropin-Releasing Factor Signaling Using DREADDs Reduces Binge- Like Ethanol Consumption of CRF-Cre Transgenic Mice

Similar documents
THE ROLE OF CORTICOTROPIN-RELEASING FACTOR (CRF) SIGNALING IN THE VENTRAL TEGMENTAL AREA IN THE REGULATION OF BINGE-LIKE ALCOHOL CONSUMPTION

Repeated Cycles of Binge-Like Ethanol Drinking in C57BL/6J Mice Augments Subsequent Voluntary Ethanol Intake But Not Other Dependence-Like Phenotypes

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

Deficits in amygdaloid camp-responsive element binding protein signaling play a role in genetic predisposition to anxiety and alcoholism

Dayna M. Hayes, Ph.D.

DEGREE (if applicable)

Adolescent Prozac Exposure Enhances Sensitivity to Cocaine in Adulthood INTRODUCTION

Adolescent Binge Drinking Leads to Changes in Alcohol Drinking, Anxiety, and Amygdalar Corticotropin Releasing Factor Cells in Adulthood in Male Rats

Jonathan Kindberg BNF Molecular Biology Through Discovery. Research Proposal

Running head: EFFECTS OF ALCOHOL ON OBJECT RECOGNITION. Impairing Effects of Alcohol on Object Recognition. A Senior Honors Thesis

Supplementary Methods. the ventrolateral orbitofrontal cortex (VLO) and basolateral amygdala (BLA). AAV8-CaMKII-HAhM

LAUREN C. ANDERSON Boston College, Department of Psychology Chestnut Hill, MA

J.C. Crabbe et al. BIOL PSYCHIATRY 2009;65: dark phase, when they are normally engaged in their highest levels of eating, drinking, and act

Neurobiology of Addiction JeanAnne Johnson Talbert, DHA, APRN BC, FNP, CARN AP

Mk-801 Administration in Adolescent Male Rats and Cocaine Conditioned Place

Drug and Alcohol Dependence and Withdrawal Models

Genetic Contributors to Alcohol Use and Misuse in Young People

How to measure rodent behavior and perform a neurological screen.

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

processes in the central nervous system (CNS), affecting many of the during the course of ethanol treatment. Ethanol stimulates the release of

RUNNING HEAD: HPA AXIS ACTIVATION BY ETHANOL DEPENDENCE 1. HPA Axis Activation by Ethanol Dependence in Adult and Adolescent Rats.

NIH Public Access Author Manuscript Alcohol. Author manuscript; available in PMC 2015 December 01.

Relationship Between Stress and Substance Use Disorders: Neurobiologic Interface

NPY Signaling Inhibits Extended Amygdala CRF Neurons to Suppress Binge Alcohol Drinking

Cognitive Function Test. NOR & spatial NOR task. Introduction. Novel Object Recognition (NOR) Estrogen Receptor (ER) in Brain

HHS Public Access Author manuscript Alcohol Clin Exp Res. Author manuscript; available in PMC 2016 April 01.

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

Hypothalamic-specific proopiomelanocortin deficiency reduces alcohol drinking in male and female mice

KRISTEN E. PLEIL, Ph.D.

The effects of environmental enrichment on nicotine sensitization in a rodent model of schizophrenia

Nicotine Decreases Ethanol-induced Dopamine Signaling and Increases Self-administration via Steroid Hormones

Zhu et al, page 1. Supplementary Figures

Supporting Online Material for

Understanding Addiction and Its Impact on the Brain. SDSMA Webinar Matthew Stanley, DO

Chelsea Kasten, PhD. Postdoctoral Fellow LSU Health Sciences Center New Orleans

Developmental regulation of Medium Spiny Neuron dendritic arborization. Lorene M. Lanier Department of Neuroscience

GABAergic Influences Increase Ingestion across All Taste Categories. Liz Miller. Molly McGinnis. Lindsey Richardson

Food restriction: enhancing effects on drug reward and striatal cell signaling

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

MeCP2 and psychostimulantinduced behavioral adaptations. Anne E. West, M.D., Ph.D. Department of Neurobiology Duke University Medical Center

Supplementary Figure 1

Effects of Systemic Administration of 8-OH-DPAT on Agonistic Social Behaviors in Male Syrian Hamsters

LESIONS OF THE MESOLIMBIC DOPAMINE SYSTEM DISRUPT SIGNALLED ESCAPE RESPONSES IN THE RAT

C81ADD Psychology of Addiction. Alcohol. Ethyl alcohol (ethanol) School of Psychology. Tobias Bast.

The Effects of Chemotherapy on Cognitive Behavior and Neurogenesis in an Animal Model of Pre- and Post- Menopausal Females

Novel Compounds for Treatment of Alcohol Addiction and Anxiety

SUPPLEMENTARY INFORMATION

NEUROBIOLOGY ALCOHOLISM

Disclosures for Prof D L Hay. Research funding: Alder Biopharmaceuticals Inc. Consulting arrangements: Intarcia Therapeutics Inc.

SUPPLEMENTARY INFORMATION

Getting into the weed: the endocannabinoid system of the gut-brain axis in energy homeostasis. Keith Sharkey

Adolescent Brain Development and Drug Abuse. Ken C. Winters, Ph.D. Professor, Department of Psychiatry, University of Minnesota

Vol 6, Iss 9, May 5, 2016

MANAGING PAIN IN PATIENTS WITH SUBSTANCE USE DISORDER

Evaluation of Anxiolytic Effect of XXX in Guinea Pig Maternal Separation Test.

Preclinical Psychopharmacology: From Mechanisms & Molecules to Medicines

At a Glance. Background Information. Lesson 3 Drugs Change the Way Neurons Communicate

Methamphetamine-Induced Conditioned Place Preference In Adolescent Male and Female Mice of Two Strains

AN EXPERIMENTAL MODEL OF ALCOHOL-INDUCED ANXIETY AND DEPRESSIVE BEHAVIOUR IN RATS

Seizure Sensitivity and GABAergic Modulation of Ethanol Sensitivity in Selectively Bred FAST and SLOW Mouse Lines 1

Orbitofrontal and striatal circuits dynamically encode the shift between goal-directed and habitual actions

The Nervous System Mark Stanford, Ph.D.

ECHO Presentation Addiction & Brain Function

SUPPLEMENTARY INFORMATION

Subjects. Thirty-five adult male Lister Hooded rats (Charles River, Kent, UK),

DREADD Targeting Projections from the Nucleus Accumbens to the Ventral Pallidum with Nicotine Self-Administration

<student name> Undergraduate Research Grant Proposal

S100B+ Cell Density day Spine Formation (%)

Chemogenetic manipulation of ventral pallidal neurons impairs acquisition of sign-tracking in rats

The Neuroscience of Addiction: A mini-review

Dopaminergic dynamics underlying sex-specific cocaine reward

Nature Neuroscience: doi: /nn Supplementary Figure 1

Basal Ganglia Anatomy, Physiology, and Function. NS201c

HIV and alcohol use: why is risk reduction in alcohol use important in HIV care?

The CRF-1 receptor antagonist, CP-154,526, attenuates stress-induced increases in ethanol consumption by BALB/cJ mice. Emily Geyer Lowery

10/27/09! Psychopharmacology 101 for the LADC! Disclosures! Lecture outline! Structural divisions of the nervous system!

Copyright 2017 The Guilford Press

Learning outcomes: Keeping it Real and Safe: What Every School Counselor Should Know About Underage Drinking

Does nicotine alter what is learned about non-drug incentives?

Running head: ENVIRONMENTAL ENRICHMENT AND ALCOHOL ADDICTION 1

Psychology in Your Life

Binge eating is a pathological feeding pattern affecting 4.5% of Americans over their

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2

Social transfer of pain in mice

Emotion I: General concepts, fear and anxiety

A. Personal Statement

NIH Public Access Author Manuscript Nat Neurosci. Author manuscript; available in PMC 2006 September 5.

Sensory Processing SensiTivity AND drug Use recovery Pathways STANDUP. Judith Homberg (NL) Fabio Fumagalli (IT) Maria Melchior (FR) Boris Quednow (CH)

Behavioral implications of the dynamic regulation of microglia: the influence of a day versus a lifetime

Ligand-Gated Ion Channels

Oxytocin and Early Experience. Sue Carter The Brain Body Center Department of Psychiatry University of Illinois at Chicago

The future of pharmacological treatment.

Supplementary Figure 1

Supplemental Data. Epithelial-Macrophage Interactions Determine Pulmonary Fibrosis Susceptibility in. Hermansky-Pudlak Syndrome

9.98 Neuropharmacology January (IAP) 2009

TEEN VULNERABILITY: DRUG EXPOSURE DURING ADOLESCENCE HAS LONG-LASTING CONSEQUENCES

Nature Neuroscience: doi: /nn Supplementary Figure 1. ACx plasticity is required for fear conditioning.

Reversing the Effects of Fragile X Syndrome

Neurobiology of Addiction

Transcription:

Manipulation of Corticotropin-Releasing Factor Signaling Using DREADDs Reduces Binge- Like Ethanol Consumption of CRF-Cre Transgenic Mice By Suzahn Ebert Senior Honors Thesis Department of Biology University of North Carolina at Chapel Hill March 24, 2016 This thesis has been prepared in conjunction with BIOL692H and in partial satisfaction of the requirements for graduation from UNC-CH with research honors. Approved: S. Alex Marshall, Ph.D., Thesis Advisor Steve Crews, Ph.D., Reader Amy Schaub Maddox, Ph.D., Reader

Abstract Binge-like ethanol consumption is characterized by the rapid achievement of blood ethanol concentrations (BEC) of 80 mg/dl and is associated with an increased risk of ethanol dependence. Evidence of this association is based on an overlap between neurobiological mechanisms active during ethanol dependence and those activated during excessive ethanol consumption prior to dependence. One such contributor is the corticotropin-releasing factor (CRF) system. Our laboratory has shown that antagonism of CRF receptors within the central amygdala (CeA) reduces binge-like ethanol consumption. The current study was designed to extend previous research by measuring ethanol consumption following the manipulation of CRF signaling in the CeA using Cre-induced inhibitory designer receptors exclusively activated by designer drugs (DREADDs) technology. Additionally, we tested whether manipulation of CRF signaling in the CeA modulates general rewarding behavior or specifically reduces binge-like ethanol consumption. Ethanol or sucrose, consumption of which is indicative of general rewarding behavior, was administered using a 4-day procedure that mimics binge-like drinking. On the first two test days, mice received drug injections to activate the DREADDs, thereby inhibiting CRF neurons within the CeA. Statistical analysis revealed that Gi-DREADD activation in the CeA reduced ethanol consumption but had no significant effect on sucrose consumption. These data suggest that the reduced ethanol consumption during the first two cycles was not merely a general rewarding behavior but was instead specific to ethanol consumption. Moreover, they suggest that inhibition of the CRF signaling pathway in the CeA remains a viable target for manipulating binge-like ethanol consumption. Introduction Binge drinking is a destructive pattern of heavy ethanol consumption characterized by the rapid achievement of blood ethanol concentrations (BECs) greater than 80 mg/dl (Council, 2004). Research indicates that binge drinking is associated with an increased risk of developing alcohol dependence, a severe yet common health concern (Jennison, 2004; McCarty et al., 2004). In its earliest stages, research on the neurobiological effects of binge-like ethanol consumption relied on rodent models that involved

voluntary consumption (McBride, 1998). However, rodents do not voluntarily consume enough ethanol under standard conditions to produce pharmacologically relevant BECs or to generate behavioral intoxication (Spanagel, 2000), both of which characterize human binge drinking. Rodent models have since been developed to more accurately model human binge drinking. One such model is the Drinking in the Dark (DID) paradigm which regularly produces BECs greater than 80 mg/dl and evidence of behavioral intoxication (Rhodes et al., 2005). Thus, the DID model simultaneously serves as a rodent correlate of human binge drinking as well as a useful tool for studying the neurobiological mechanisms that underlie binge-like ethanol consumption in mice (Sprow and Thiele, 2012). Neurobiological mechanisms involved in excessive ethanol consumption prior to dependence overlap with those associated with ethanol dependence (Sprow and Thiele, 2012); thus, studies investigating the neurobiological systems that underlie binge ethanol consumption have the potential to shed light on the neurobiological changes that precede alcohol dependence. One such system is the corticotropin-releasing factor (CRF) system, which is implicated both in the regulation of ethanol dependence and excessive consumption. CRF is an amino acid peptide present in a wide variety of cell types, including neurons. CRF is most commonly associated with the general mammalian stress response, but considering the relationship between stress and alcohol consumption, it is not surprising that CRF signaling modulates ethanol consumption (Martin et al., 2010; Bahi, 2013). Previous research has illustrated that CRF plays a key role in excessive, dependence-like ethanol consumption in rats through the activation of G-protein coupled CRF-1 receptors (CRF1R) in the central nucleus of the amygdala (CeA) (Roberto et al., 2010). The CeA is known to play a fundamental role in physiological and behavioral responses to drug-related stimuli, and neuroadaptations within the CeA have been shown to play a central role in alcohol dependence (Gilpin, 2012). Our laboratory has built upon the aforementioned studies involving the CRF system in the CeA by showing that not only does ethanol dysregulate

the CRF system, but also that antagonism of CRF receptors within the CeA reduces binge-like ethanol consumption (Lowery et al., 2010). The current study extends previous research by measuring ethanol consumption following the manipulation of CRF signaling in the CeA using Cre-induced Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) technology. DREADDs are G-protein coupled receptors which are activated by small, drug-like molecules. They are particularly useful as a means of investigating and manipulating discrete populations of neurons in the brain, as they have reduced off-target effects and heightened functional selectivity compared to other psychopharmological methods, including agonist and antagonist ligands (Fortress et al., 2015; Lee and Roth, 2014). For this study, the Gi-DREADD, which induces neuronal silencing, was used to selectively inhibit CRF signaling in the CeA. Materials and Methods Animals CRF-Cre transgenic mice, backcrossed with C57s, were bred in house and were 10-18 weeks of age at the onset of the study. These mice expressed the exogenous Cre-recombinase only in cells that expressed endogenous CRF (Martin et al., 2010). All mice were housed in individual plastic cages with constant access to Purina RMH 3000 chow. Water was available at all times except when the mice were given access to alcohol or sucrose. The animal colony room was kept at a temperature of approximately 22⁰C and operated on a 12 hour light: 12 hour dark cycle (lights on at 08:00 hours). All procedures were performed in accordance with the National Institute of Health Guidelines for the Care and Use of Laboratory Animals and were approved by the University of North Carolina Institutional Animal Care and Use Committee (National Research Council, 2011). Stereotactic Surgery and DREADDS

The study used surgically delivered Cre-inducible adeno-associated viruses to encode for designer receptors exclusively activated by designer drugs (DREADDs) as a means of investigating specific neurological changes associated with binge-like alcohol consumption (Fortress et al., 2015). The DREADDs were incorporated into the viral genome, so the neural cells of interest were exposed to the virus and subsequently expressed the designer receptor. Prior to surgery, mice received an anesthetic dose of a ketamine cocktail as well as a topical anesthetic. Stereotactic surgery was used to bilaterally infuse either the inhibitory Gi-DREADD (AAV8-hSyn-DIO-hM4D-mCherry) or a control virus with a fluorescent tag (AAV8-hSyn-DIO-mCherry) at the following coordinates in the central amygdala: AP: -1.05 ML:+/-2.5 DV:-4.64. A Hamilton syringe was used to inject a total of 0.3 µl bilaterally at a rate of 0.1 µl per minute. After each injection, the virus was allowed to diffuse for ten minutes. Drinking-in-the-Dark Procedure Ethanol was administered using the DID procedure, a well-established 4-day binge-like drinking model (Rhodes et al., 2005, 2007; Sparta et al., 2008; Lowery et al., 2010). Animals were given access to a 20% (v/v) ethanol solution three hours into the dark cycle on each of the 4 days. Tap water and 95% ethyl alcohol were used to prepare the 20% (v/v) ethanol solution. The ethanol was available for a period of two hours on days 1-3. Day 4 was classified as the binge test day, and

the ethanol was available for 4 hours. Animals underwent 3 consecutive weeks of ethanol testing followed by an open field test and one week of sucrose testing. Sucrose was administered using the DID procedure and served as a control to examine the specificity of the inhibition of CRF neurons to ethanol consumption. Immediately preceding the DID procedures in week 1 and 2 of the ethanol test and the sucrose test, mice were injected intraperitoneally with 0.1 mg/kg clozapine-n-oxide (CNO) to induce Gi activation which in turn induced neuronal silencing in CRF neurons of the CeA expressing the DREADDs. However, immediately preceding week 3 of ethanol testing, mice were injected with saline instead of CNO to measure ethanol consumption in the absence of manipulation of CRF signaling. Approximately 60 µl of tail blood was collected from each animal immediately following the end of the binge drinking period on day 4 of each test week. The samples were used to determine individual BECs using an Analox Analyzer (Analox Instruments). Open Field Testing Mice used in the DID tests were split into two groups, receiving either intraperitoneal CNO or saline, and subsequently subjected to open-field locomotor activity tests. The purpose of the openfield tests is to measure anxiety-like behavior, as avoidance of the central area of the chamber reflects a heightened state of anxiety (Choleris et al., 2001; Fee et al., 2004). The open field tests were conducted identical to previous reports using locomotor activity chambers made of clear Plexiglass and measuring 40.64 x 40.64 x 30.48 cm (Sparta et al., 2008). Testing sessions were 4 hours long, with measurements taken every 5 minutes. Total distance traveled (cm) was measured to assess the effect of CNO induced Gi activation in the CeA on locomotor activity, and center distance (cm) and time were used to examine the effects of silencing CeA CRF neurons on anxiety. Placement Checks At the conclusion of the study, mice were euthanized with a sodium pentobarbital overdose and transcardially perfused with 0.1M phosphate buffered saline followed by 4% paraformaldehyde.

The brains were extracted, sliced in 40 µm sections using a vibrating microtome (Leica VT1000S; Wetzlar, Germany), and mounted. The placement of the virus was visualized and confirmed to be in the CeA using an Olympus BX-51 fluorescent microscope set to a 100x magnification. Data Analysis Two tailed t-tests were performed to determine if Gi-DREADD activation had a significant effect on ethanol consumption. Two-way ANOVAs were used to determine if CNO induced Gi activation in the CeA had an effect on locomotor activity and to examine the effects of silencing CeA CRF neurons on anxiety. Results DID Trials During the first two weeks of ethanol testing, mice received CNO injections, inhibiting CRF neurons within the CeA. T-tests were used to determine if Gi-DREADD activation had a significant effect on ethanol consumption. Gi-DREADD activation in the CeA resulted in reduced ethanol consumption ([Test 1: Gi = 2.875 ± 0.3775 g/kg ethanol; control = 5.036 ± 0.3765 g/kg ethanol; p=0.0006]; [Test 2: Gi = 3.384 ± 0.2911 g/kg ethanol; control = 5.009 ± 0.4624 g/kg ethanol; p=0.0075]). Accordingly, there was also a significant reduction in BECs in test 1; however, there was no significant reduction in BECs in test 2 ([Test 1: Gi = 35.87 ± 8.71 mg/dl; control = 73.92 ± 11.68 mg/dl; p=0.0167]; [Test 2: Gi = 40.93 ± 10.49 mg/dl; control = 71.63 ± 12.82 mg/dl; p=0.0787]).

E th a n o l C o n s u m p tio n (g /k g ) Figure 1. Two tailed t-tests indicated a significant reduction in ethanol consumption upon Gi activation in the CeA for test 1 (p = 0.0006; n=11) and test 2 (p=0.0075; n=11). Figure 2. Two tailed t-tests indicated a significant reduction in BECs upon Gi activation in the CeA for test 1 (p=0.0167; n=11), but no significant reduction for test 2 (p=0.0787; n=11). To determine if CNO had an effect independent of the virus, mice received a saline injection prior to the third round of ethanol testing. No significant difference in ethanol consumption was observed between the experimental and control group under these conditions (Test 3: Gi = 4.250 ± 0.2980 g/kg ethanol; control = 4.541 ± 0.5232 g/kg ethanol; p= 0.6339). Additionally, there was no significant difference in BECs under these conditions (Test 3: Gi = 69.07 ± 13.27 mg/dl; control = 71.83 ± 10.88 mg/dl; p=0.8743). T e s t 3 S a lin e T r e a tm e n t 6 4 2 0 C o n t r o l G i

S u c r o s e C o n s u m p tio n (g /k g ) B lo o d E th a n o l C o n c e n tr a tio n (m g /d L ) Figure 3. A two tailed t-test indicated no significant difference in ethanol consumption in mice treated with saline (p= 0.6339; n=10). T e s t 3 S a lin e T r e a tm e n t 1 0 0 8 0 6 0 4 0 2 0 0 C o n t r o l G i Figure 4. A two tailed t-test indicated no significant difference in BECs in mice treated with saline (p=0.8743; n=10). In the final round of testing, mice were given access to sucrose solution in the place of ethanol to test the specificity of Gi DREADD manipulations in the CeA and to determine if manipulating CRF signaling had an effect on general rewarding tendencies. A t-test showed no significant effect of Gi DREADD activation on sucrose consumption (Test 4: Gi = 8.997 ± 1.503 g/kg sucrose; control = 8.333 ± 1.225 g/kg sucrose; p= 0.7436). T e s t 4 C N O T r e a tm e n t 1 2 8 4 0 C o n t r o l G i Figure 5. A two tailed t-test indicated no significant difference in sucrose consumption upon Gi activation in the CeA (p= 0.7436; n=11).

T o ta l D is ta n c e (c m ) Open Field Tests Mice received either an intraperitoneal CNO or saline injection and were subjected to openfield locomotor activity tests. Total distance traveled was measured to assess the effect of CNO induced Gi activation in the CeA on locomotor activity, and center distance and time were used to examine the effects of silencing CeA CRF neurons on anxiety. For center distance and total distance, a main effect of time was observed [(Center distance: F=6.36; p<0.0001); (Total distance: F=5.80; p<0.0001)], but there was no main effect of Gi activation in the CeA [(Center distance: F=2.31; p=0.1451); (Total distance: F=0.79; p=0.3859)]. For center time, no main effect of Gi activation (F=1.35; p=0.2599) or of time was observed (F=1.01; p=0.4478). 1 5 0 0 T o ta l D is ta n c e -C o m b in e d 1 0 0 0 G i C o n tro l 5 0 0 0 1 0 2 0 3 0 4 0 5 0 6 0 7 0 8 0 9 0 1 0 0 1 1 0 1 2 0 1 3 0 1 4 0 1 5 0 1 6 0 1 7 0 1 8 0 1 9 0 2 0 0 2 1 0 2 2 0 2 3 0 2 4 0 T im e (m in s ) Figure 6. A two way ANOVA indicated a main effect of time but no significant interaction or main effect of Gi activation in the CeA in total distance.

C e n te r D is ta n c e (c m ) C e n te r D is ta n c e -C o m b in e d 6 0 0 4 0 0 G i C o n tro l 2 0 0 0 1 0 2 0 3 0 4 0 5 0 6 0 7 0 8 0 9 0 1 0 0 1 1 0 1 2 0 1 3 0 1 4 0 1 5 0 1 6 0 1 7 0 1 8 0 1 9 0 2 0 0 2 1 0 2 2 0 2 3 0 2 4 0 T im e (m in s ) Figure 7. A two way ANOVA indicated a main effect of time but no significant interaction or main effect of Gi activation in the CeA in center distance.

T im e S p e n t In C e n te r A re n a (s e c s ) C e n t e r T im e -C o m b in e d 1 5 0 1 0 0 5 0 G i C o n tro l 0 1 0 2 0 3 0 4 0 5 0 6 0 7 0 8 0 9 0 1 0 0 1 1 0 1 2 0 1 3 0 1 4 0 1 5 0 1 6 0 1 7 0 1 8 0 1 9 0 2 0 0 2 1 0 2 2 0 2 3 0 2 4 0 T im e (m in s ) Figure 8. No interaction or main effects were observed in center time. Discussion The current study confirms previous findings that modulation of the CRF system in the CeA plays a role in alcohol consumption. The results showed that CRF neuronal inhibition in the CeA using Cre-induced DREADD technology reduced binge-like ethanol consumption. There was no significant difference in ethanol consumption in mice treated with saline, indicating that CNO did not have an effect independent of the virus. Additionally, there was no significant effect of Gi DREADD activation on sucrose consumption, suggesting that the effects of CRF receptor antagonism in the CeA is specific to ethanol consumption and does not affect general rewarding tendencies. The results of the open field tests indicated that CRF receptor antagonism in the CeA had no effect on locomotor activity or anxiety. Together, the results of the DID tests and the open field tests suggest that inhibition of the CRF signaling pathway in the CeA specifically, reduces binge-like

ethanol consumption without decreasing activity or increasing anxiety. Thus, it remains a viable target for manipulating binge-like ethanol consumption without other negative consequences. The mounting evidence for the role of CRF in excessive, binge-like ethanol consumption suggests an overlap between the neurobiological systems involved in the general mammalian stress response and those involved in the regulation of excessive alcohol consumption and dependence. The fundamental role of CRF receptors in the regulation of stress and alcohol dependence is welldocumented, but recent studies have shown that peptides other than CRF, including a variety of urocortins, may also act on CRF receptors to modulate both stress and alcohol consumption (Ryabinin et al., 2012). Among others, the aforementioned studies demonstrate that stress is a risk factor for alcoholism. Thus, the neurobiological relationship between stress and alcohol consumption merits further study, as it could provide new insight into why people drink, how alcohol affects their behavior, and why some people become dependent on alcohol. The current findings are limited to the role of CRF in the CeA, so it would be interesting to investigate the potential of manipulating CRF signaling in other regions of the amygdala to influence ethanol consumption. Specifically, the basal lateral amygdala (BLA) is known to have glutamatergic projections to the CeA (Ren et al., 2010), which could allow for upstream targeting of the CRF system. In conclusion, the current study sheds light on the role of the CRF signaling in the CeA in ethanol consumption. On a broader scale, it implicates CRF signaling in other parts of the brain as well as additional signaling pathways involved in the stress response in the regulation of excessive alcohol consumption. Acknowledgements The authors thank S. Alex Marshall and Todd E. Thiele for their guidance and support, as well as Rhiannon D. Thomas, Timothy P. Gilliam, and Sonia Sabater for their technical assistance. Funding and Disclosure

The author declares no conflict of interest. This work was supported by National Institutes of Health grants AA022048, AA013573, AA015148, AA021611, AA011605, & GM000678. References 1. Bahi A. Increased anxiety, voluntary alcohol consumption and ethanol-induced place preference in mice following chronic psychosocial stress. Stress. 2013;16(4):441-451. 2. Choleris E, Thomas AW, Kavaliers M, Prato FS. A detailed ethological analysis of the mouse open field test: effects of diazepam, chlordiazepoxide and an extremely low frequency pulsed magnetic field. Neuroscience & Biobehavioral Reviews. 2001;25(3):235-260. 3. Council NIAAA. NIAAA council approves definition of binge drinking. NIAAA Newsletter; 2004. 4. Fee JR, Sparta DR, Knapp DJ, Breese GR, Picker MJ, Thiele TE. Predictors of High Ethanol Consumption in RIIβ Knock-Out Mice: Assessment of Anxiety and Ethanol- Induced Sedation. Alcoholism, clinical and experimental research. 2004;28(10):1459-1468. 5. Fortress AM, Hamlett ED, Vazey EM, et al. Designer Receptors Enhance Memory in a Mouse Model of Down Syndrome. The Journal of Neuroscience. 2015;35(4):1343-1353. 6. Gilpin NW. Neuropeptides in Central Amygdala: Role in Anxiety- and Alcohol-Related Behaviors. Alcohol (Fayetteville, N.Y.). 2012;46(4):329-337. 7. Jennison KM. The Short Term Effects and Unintended Long Term Consequences of Binge Drinking in College: A 10 Year Follow Up Study. The American Journal of Drug and Alcohol Abuse. 2004;30(3):659-684.

8. Lee H-M, Giguere PM, Roth BL. DREADDs: novel tools for drug discovery and development. Drug discovery today. 2014;19(4):469-473. 9. Lowery EG, Spanos M, Navarro M, Lyons AM, Hodge CW, Thiele TE. CRF-1 Antagonist and CRF-2 Agonist Decrease Binge-Like Ethanol Drinking in C57BL/6J Mice Independent of the HPA Axis. Neuropsychopharmacology. 2010;35(6):1241-1252. 10. Martin EI, Ressler KJ, Jasnow AM, et al. A novel transgenic mouse for gene-targeting within cells that express corticotropin-releasing factor. Biological psychiatry. 2010;67(12):1212-1216. 11. McBride WJW. Animal models of alcoholism: neurobiology of high alcohol-drinking behavior in rodents. Critical reviews in neurobiology. 1998;12(4):339-369. 12. McCarty CA, Ebel BE, Garrison MM, DiGiuseppe DL, Christakis DA, Rivara FP. Continuity of Binge and Harmful Drinking From Late Adolescence to Early Adulthood. Pediatrics. 2004;114(3):714-719. 13. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th edition. Washington (DC): National Academies Press (US); 2011. Available from: http://www.ncbi.nlm.nih.gov/books/nbk54050/ 14. Ren W, Neugebauer V. Pain-related increase of excitatory transmission and decrease of inhibitory transmission in the central nucleus of the amygdala are mediated by mglur1. Molecular Pain. 2010;6:93-93. 15. Rhodes JS, Best K, Belknap JK, Finn DA, Crabbe JC. Evaluation of a simple model of ethanol drinking to intoxication in C57BL/6J mice. Physiology & Behavior. 2005;84(1):53-63.

16. Rhodes JS, Ford MM, Yu CH, et al. Mouse inbred strain differences in ethanol drinking to intoxication. Genes, Brain and Behavior. 2007;6(1):1-18. 17. Roberto M, Cruz MT, Gilpin NW, et al. Corticotropin Releasing Factor Induced Amygdala Gamma-Aminobutyric Acid Release Plays a Key Role in Alcohol Dependence. Biological psychiatry. 2010;67(9):831-839. 18. Ryabinin AE, Tsoory MM, Kozicz T, et al. Urocortins: CRF s siblings and their potential role in anxiety, depression and alcohol drinking behavior. Alcohol (Fayetteville, N.y.). 2012;46(4):349-357. 19. Spanagel, R. Recent animal models of alcoholism. Alcohol Research and Health. 2000;24(2):124-31. Retrieved from: http://libproxy.lib.unc.edu/login?url=http://search.proquest.com/docview/222452098?acc ountid=14244 20. Sparta DR, Sparrow AM, Lowery EG, Fee JR, Knapp DJ, Thiele TE. Blockade of the Corticotropin Releasing Factor Type Receptor Attenuates Elevated Ethanol Drinking Associated With Drinking in the Dark Procedures. Alcoholism, clinical and experimental research. 2008;32(2):259-265. 21. Sprow GM, Thiele TE. The Neurobiology of Binge-Like Ethanol Drinking: Evidence from Rodent Models. Physiology & behavior. 2012;106(3):325-331.