1
|
Solomon MG, Nennig SE, Cotton MR, Whiting KE, Fulenwider HD, Schank JR. Neurokinin-1 receptors in the nucleus accumbens shell influence sensitivity to social defeat stress and stress-induced alcohol consumption in male mice. ADDICTION NEUROSCIENCE 2024; 13:100174. [PMID: 39801674 PMCID: PMC11720327 DOI: 10.1016/j.addicn.2024.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Chronic social defeat stress (SDS) is a widely employed preclinical model of depression involving repeated exposure to physical defeats using a resident-intruder model in male mice. Exposure to SDS induces depressive-like phenotypes including anhedonia, social withdrawal, and increased drug and alcohol consumption. Previously, we found that expression of the neurokinin-1 receptor (NK1R) is increased in the nucleus accumbens (NAC) of mice that are sensitive to this stressor and increase their alcohol intake. The NK1R is the endogenous receptor for the neuropeptide substance P (SP) and plays a prominent role in stress, anxiety, and addiction. In the present study, we assessed changes in NK1R protein levels in the NAC shell and implemented viral vector strategies to demonstrate a functional role of the NK1R in sensitivity to SDS. Specifically, we found that NK1R protein levels were increased in the NAC shell following SDS exposure. Next, we found that NK1R overexpression in the NAC shell increased the sensitivity to SDS and stress-induced alcohol consumption. Together, these experiments provide evidence for a role of the NK1R in the NAC shell in the sensitivity to SDS and the subsequent escalation in alcohol intake.
Collapse
Affiliation(s)
- Matthew G Solomon
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Sadie E Nennig
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Mallory R Cotton
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Kimberly E Whiting
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Hannah D Fulenwider
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Jesse R Schank
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| |
Collapse
|
2
|
Meinhardt MW, Gerlach B, Spanagel R. Good Practice Guideline for Preclinical Alcohol Research: The STRINGENCY Framework. Curr Top Behav Neurosci 2024. [PMID: 39117860 DOI: 10.1007/7854_2024_484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Research in the field of preclinical alcohol research, but also science in general, has a problem: Many published scientific results cannot be repeated. As a result, findings from preclinical research often do not translate well to humans, causing increasing disappointment and calls for restructuring of preclinical research, that is, better reproducibility of preclinical research. However, the replication crisis is an inherent problem in biomedical research. Replication failures are not only due to small experimental variations but are often the result of poor methodology. In response to the replication crisis, numerous guidelines and recommendations have been proposed to promote transparency, rigor, and reproducibility in scientific research. What is missing today is a framework that integrates all the confusing information that results from all these guidelines and recommendations. Here we present STRINGENCY, an integrative approach to good practice guidelines for preclinical alcohol research, which can also apply to behavioral research in general and which aims to improve preclinical research to better prepare it for translation and minimize the "valley of death" in translational research. STRINGENCY includes systematic review and, when possible, meta-analysis prior to study design, sample size calculation, preregistration, multisite experiments, scientific data management (FAIR), reporting of data using ARRIVE, generalization of research data, and transparent publications that allow reporting of null results. We invite the scientific community to adopt STRINGENCY to improve the reliability and impact of preclinical alcohol research.
Collapse
Affiliation(s)
- Marcus W Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.
| | - Björn Gerlach
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Guarantors of EQIPD e.V., Heidelberg, Germany
- PAASP GmbH, Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- German Center for Mental Health (DZPG), Mannheim, Heidelberg, Ulm, Germany.
| |
Collapse
|
3
|
Scopano MR, Jones HE, Stea SG, Freeman MZ, Grisel JE. Age, β-endorphin, and sex dependent effects of maternal separation on locomotor activity, anxiety-like behavior, and alcohol reward. Front Behav Neurosci 2023; 17:1155647. [PMID: 37091593 PMCID: PMC10113444 DOI: 10.3389/fnbeh.2023.1155647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/07/2023] [Indexed: 04/09/2023] Open
Abstract
IntroductionChildhood adversity is pervasive and linked to numerous disadvantages in adulthood, including physical health problems, mental illness, and substance use disorders. Initial sensitivity to the rewarding effects of alcohol predicts the risk of developing an alcohol use disorder, and may be linked to developmental stress. The opioid peptide β-endorphin (β-E) regulates the stress response and is also implicated in the risk for excessive alcohol consumption.MethodsWe explored the influence of β-E in an animal model of early life adversity using controlled maternal separation by evaluating changes in locomotor activity, anxiety-like behavior, and the initial rewarding effects of alcohol in a single exposure conditioned place preference paradigm in control C57BL/6J and β-E deficient β-E +/+ 0.129S2-Pomc tm1Low/J; β-E −/− mice. Maternal separation (MS) occurred for 3 h each day from post-natal days (PND) 5–18 in approximately half the subjects.ResultsMaternal interactions increased following the separation protocol equally in both genotypes. MS and control subjects were tested as adolescents (PND 26–32) or adults (PND 58–72); the effects of MS were generally more pronounced in older subjects. Adults were more active than adolescents in the open field, and MS decreased activity in adolescent mice but increased it in adults. The increase in adult activity as a result of early life stress depended on both β-E and sex. β-E also influenced the effect of maternal separation on anxiety-like behavior in the Elevated Plus Maze. MS promoted rewarding effects of alcohol in male β-E deficient mice of either age, but had no effect in other groups.DiscussionTaken together, these results suggest that the effects of MS develop over time and are β-E and sex dependent and may aid understanding of how individual differences influence the impact of adverse childhood experiences.
Collapse
|
4
|
Mantsch JR. Corticotropin releasing factor and drug seeking in substance use disorders: Preclinical evidence and translational limitations. ADDICTION NEUROSCIENCE 2022; 4:100038. [PMID: 36531188 PMCID: PMC9757758 DOI: 10.1016/j.addicn.2022.100038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The neuropeptide, corticotropin releasing factor (CRF), has been an enigmatic target for the development of medications aimed at treating stress-related disorders. Despite a large body of evidence from preclinical studies in rodents demonstrating that CRF receptor antagonists prevent stressor-induced drug seeking, medications targeting the CRF-R1 have failed in clinical trials. Here, we provide an overview of the abundant findings from preclinical rodent studies suggesting that CRF signaling is involved in stressor-induced relapse. The scientific literature that has defined the receptors, mechanisms and neurocircuits through which CRF contributes to stressor-induced reinstatement of drug seeking following self-administration and conditioned place preference in rodents is reviewed. Evidence that CRF signaling is recruited with repeated drug use in a manner that heightens susceptibility to stressor-induced drug seeking in rodents is presented. Factors that may determine the influence of CRF signaling in substance use disorders, including developmental windows, biological sex, and genetics are examined. Finally, we discuss the translational failure of medications targeting CRF signaling as interventions for substance use disorders and other stress-related conditions. We conclude that new perspectives and research directions are needed to unravel the mysterious role of CRF in substance use disorders.
Collapse
Affiliation(s)
- John R Mantsch
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, United States
| |
Collapse
|
5
|
Excessive alcohol consumption after exposure to two types of chronic social stress: intermittent episodes vs. continuous exposure in C57BL/6J mice with a history of drinking. Psychopharmacology (Berl) 2022; 239:3287-3296. [PMID: 35974246 DOI: 10.1007/s00213-022-06211-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/01/2022] [Indexed: 10/15/2022]
Abstract
RATIONALE The attraction to alcohol can be greatly increased when it is consumed in a social context. While pro-social interactions can potentiate voluntary alcohol drinking under some conditions, aversive social experience (i.e., social stress) can similarly intensify alcohol consumption. OBJECTIVE We sought to determine how exposure to different types of chronic social stress (i.e., intermittent episodes of social defeat or continuous social stress) influences alcohol consumption and the reinforcing effects of alcohol in mice with a history of drinking. METHODS Separate cohorts of male C57BL/6J mice were exposed to either 10 days of continuous or intermittent social defeat stress. In experiment 1, mice were assigned to 20% w/v alcohol consumption in a two-bottle choice protocol both prior to and after exposure to social defeat stress. In a second experiment, mice engaged in an operant response sequence to gain access to alcohol wherein completion of a fixed interval (FI; 5 min) schedule was reinforced with continuous access to alcohol (fixed ratio; FR1) for up to 1.8 g/kg. Alcohol-reinforced responding and subsequent alcohol consumption were assessed daily for 4 weeks prior to the 10-day social stress exposure and for 6-week post-stress. Machine learning was implemented to standardize the analysis of defeat behaviors exhibited by the intruder mouse during confrontation with an attacking resident. RESULTS In mice with a prior history of alcohol drinking, intermittent episodes of social defeat stress produced a significant increase in 20% EtOH consumption in preference over concurrently available water. This increased intake persisted for at least 6 weeks after the final social stress experience. Intermittently stressed mice also accelerated their anticipatory responding during the fixed interval component of the operant response chain that was reinforced by alcohol. Neither unstressed controls nor mice exposed to continuous social stress exhibited significant increases in alcohol consumption and alcohol reinforcement. DISCUSSION Episodic social defeat stress promotes the seeking and consumption of alcohol, extending earlier work to alcohol-experienced mice. We hypothesize that intermittent access to alcohol and intermittent episodes of social stress are additive and share common sensitizing neural mechanisms that engender excessive alcohol consumption.
Collapse
|
6
|
Mineur YS, Garcia-Rivas V, Thomas MA, Soares AR, McKee SA, Picciotto MR. Sex differences in stress-induced alcohol intake: a review of preclinical studies focused on amygdala and inflammatory pathways. Psychopharmacology (Berl) 2022; 239:2041-2061. [PMID: 35359158 PMCID: PMC9704113 DOI: 10.1007/s00213-022-06120-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023]
Abstract
Clinical studies suggest that women are more likely than men to relapse to alcohol drinking in response to stress; however, the mechanisms underlying this sex difference are not well understood. A number of preclinical behavioral models have been used to study stress-induced alcohol intake. Here, we review paradigms used to study effects of stress on alcohol intake in rodents, focusing on findings relevant to sex differences. To date, studies of sex differences in stress-induced alcohol drinking have been somewhat limited; however, there is evidence that amygdala-centered circuits contribute to effects of stress on alcohol seeking. In addition, we present an overview of inflammatory pathways leading to microglial activation that may contribute to alcohol-dependent behaviors. We propose that sex differences in neuronal function and inflammatory signaling in circuits centered on the amygdala are involved in sex-dependent effects on stress-induced alcohol seeking and suggest that this is an important area for future studies.
Collapse
Affiliation(s)
- Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee A Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Sherry A McKee
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
| |
Collapse
|
7
|
Spanagel R. Ten Points to Improve Reproducibility and Translation of Animal Research. Front Behav Neurosci 2022; 16:869511. [PMID: 35530730 PMCID: PMC9070052 DOI: 10.3389/fnbeh.2022.869511] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022] Open
Abstract
Findings from animal experiments are often difficult to transfer to humans. In this perspective article I discuss two questions. First, why are the results of animal experiments often so difficult to transfer to humans? And second, what can be done to improve translation from animal experiments to humans? Translation failures are often the result of poor methodology. It is not merely the fact that low statistical power of basic and preclinical studies undermine a "real effect," but the accuracy with which data from animal studies are collected and described, and the resulting robustness of the data is generally very low and often does not allow translation to a much more heterogeneous human condition. Equally important is the fact that the vast majority of publications in the biomedical field in the last few decades have reported positive findings and have thus generated a knowledge bias. Further contributions to reproducibility and translation failures are discussed in this paper, and 10 points of recommendation to improve reproducibility and translation are outlined. These recommendations are: (i) prior to planning an actual study, a systematic review or potential preclinical meta-analysis should be considered. (ii) An a priori power calculation should be carried out. (iii) The experimental study protocol should be pre-registered. (iv) The execution of the study should be in accordance with the most recent ARRIVE guidelines. (v) When planning the study, the generalizability of the data to be collected should also be considered (e.g., sex or age differences). (vi) "Method-hopping" should be avoided, meaning that it is not necessary to use the most advanced technology but rather to have the applied methodology under control. (vii) National or international networks should be considered to carry out multicenter preclinical studies or to obtain convergent evidence. (viii) Animal models that capture DSM-5 or ICD-11 criteria should be considered in the context of research on psychiatric disorders. (ix) Raw data of publication should be made publicly available and should be in accordance with the FAIR Guiding Principles for scientific data management. (x) Finally, negative findings should be published to counteract publication bias. The application of these 10 points of recommendation, especially for preclinical confirmatory studies but also to some degree for exploratory studies, will ultimately improve the reproducibility and translation of animal research.
Collapse
Affiliation(s)
- Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
8
|
Miczek KA, DiLeo A, Newman EL, Akdilek N, Covington HE. Neurobiological Bases of Alcohol Consumption After Social Stress. Curr Top Behav Neurosci 2022; 54:245-281. [PMID: 34964935 PMCID: PMC9698769 DOI: 10.1007/7854_2021_273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The urge to seek and consume excessive alcohol is intensified by prior experiences with social stress, and this cascade can be modeled under systematically controlled laboratory conditions in rodents and non-human primates. Adaptive coping with intermittent episodes of social defeat stress often transitions to maladaptive responses to traumatic continuous stress, and alcohol consumption may become part of coping responses. At the circuit level, the neural pathways subserving stress coping intersect with those for alcohol consumption. Increasingly discrete regions and connections within the prefrontal cortex, the ventral and dorsal striatum, thalamic and hypothalamic nuclei, tegmental areas as well as brain stem structures begin to be identified as critical for reacting to and coping with social stress while seeking and consuming alcohol. Several candidate molecules that modulate signals within these neural connections have been targeted in order to reduce excessive drinking and relapse. In spite of some early clinical failures, neuropeptides such as CRF, opioids, or oxytocin continue to be examined for their role in attenuating stress-escalated drinking. Recent work has focused on neural sites of action for peptides and steroids, most likely in neuroinflammatory processes as a result of interactive effects of episodic social stress and excessive alcohol seeking and drinking.
Collapse
Affiliation(s)
- Klaus A. Miczek
- Department of Psychology, Tufts University, Medford, MA, USA,Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Alyssa DiLeo
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Emily L. Newman
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Naz Akdilek
- Department of Psychology, Tufts University, Medford, MA, USA
| | | |
Collapse
|
9
|
Abstract
Pandemic management is likely to represent a global reality for years to come, but the roadmap for how to approach pandemic restrictions is as yet unclear. Of the restrictions enacted during COVID-19, among the more controversial surround alcohol. Like many infectious diseases, the principal mode of transmission for COVID-19 is direct respiration of droplets emitted during close social contact, and health officials warn that alcohol consumption may lead to decreased adherence to physical distancing guidelines. Governing bodies have acted to close bars before restaurants and have also specifically restricted alcohol sales, while at the same time those in the nightlife industry have labeled such actions unfounded and discriminatory. Complicating such debates is the lack of evidence on alcohol's effects on physical distance. In the current study we employed a randomized alcohol-administration design paired with computer-vision measures, analyzing over 20,000 proximity readings derived from video to examine the effect of alcohol consumption on physical distance during social interaction. Results indicated that alcohol caused individuals to draw significantly closer to an unfamiliar interaction partner during social exchange, reducing physical proximity at a rate with potentially important implications for public health. In contrast, alcohol had no effect on physical distance with a familiar interaction partner. Findings suggest that alcohol might act to overcome a natural caution people feel towards strangers and thus promote virus transmission between previously unconnected social groups.
Collapse
|
10
|
Jones CE, Wickham PT, Lim MM. Early life sleep disruption is a risk factor for increased ethanol drinking after acute footshock stress in prairie voles. Behav Neurosci 2020; 134:424-434. [PMID: 32700922 DOI: 10.1037/bne0000410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Early postnatal experiences are important for shaping the development of the stress response and may contribute to the later emergence of alcohol use disorders. We have previously found that early life sleep disruption impairs social development and alters GABA neurons in the brain of adult prairie voles, a socially monogamous rodent that displays natural ethanol preference in the laboratory. However, it is unclear whether these effects on social behavior are due, in part, to overall anhedonia and/or altered behavioral response to stress. To address this question, litters containing prairie vole pups were sleep disrupted by gentle cage agitation for 7 consecutive days from postnatal days (P) 14 to 21 (early life sleep disruption, or ELSD group) or allowed to sleep undisturbed (Control). Adult voles underwent a 2-bottle choice ethanol drinking procedure integrated with a single session of footshocks. Ethanol intake after footshock was measured as well as c-Fos immunoreactivity in the lateral and central amygdala. ELSD animals showed increased ethanol consumption and increased neural activity in these amygdala regions after footshock compared to control animals. There were no differences in baseline ethanol drinking prior to exposure to a stressor. These results suggest that early life sleep disruption in prairie voles does not produce anhedonia but can have long-lasting effects on stress reactivity. In addition to shaping species-typical social behavior, early life sleep may be important in the development of stress induced ethanol consumption and the activation of limbic pathways associated with stress. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
|
11
|
Simpson S, Shankar K, Kimbrough A, George O. Role of corticotropin-releasing factor in alcohol and nicotine addiction. Brain Res 2020; 1740:146850. [PMID: 32330519 DOI: 10.1016/j.brainres.2020.146850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/31/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022]
Abstract
The two most prevalent substance use disorders involve alcohol and nicotine, which are often co-abused. Robust preclinical and translational evidence indicates that individuals initiate drug use for the acute rewarding effects of the substance. The development of negative emotional states is key for the transition from recreational use to substance use disorders as subjects seek the substance to obtain relief from the negative emotional states of acute withdrawal and protracted abstinence. The neuropeptide corticotropin-releasing factor (CRF) is a major regulator of the brain stress system and key in the development of negative affective states. The present review examines the role of CRF in preclinical models of alcohol and nicotine abuse and explores links between CRF and anxiety-like, dysphoria-like, and other negative affective states. Finally, the present review discusses preclinical models of nicotine and alcohol use with regard to the CRF system, advances in molecular and genetic manipulations of CRF, and the importance of examining both males and females in this field of research.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States; Department of Neuroscience, Scripps Research, La Jolla, CA 92037, United States
| | - Kokila Shankar
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States; Department of Neuroscience, Scripps Research, La Jolla, CA 92037, United States
| | - Adam Kimbrough
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Olivier George
- Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.
| |
Collapse
|
12
|
Neasta J, Darcq E, Jeanblanc J, Carnicella S, Ben Hamida S. GPCR and Alcohol-Related Behaviors in Genetically Modified Mice. Neurotherapeutics 2020; 17:17-42. [PMID: 31919661 PMCID: PMC7007453 DOI: 10.1007/s13311-019-00828-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.
Collapse
Affiliation(s)
- Jérémie Neasta
- Laboratoire de Pharmacologie, Faculté de Pharmacie, University of Montpellier, 34093, Montpellier, France
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | - Jérôme Jeanblanc
- Research Group on Alcohol and Pharmacodependences-INSERM U1247, University of Picardie Jules Verne, 80025, Amiens, France
| | - Sebastien Carnicella
- INSERM U1216, Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, 38000, Grenoble, France
| | - Sami Ben Hamida
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada.
| |
Collapse
|
13
|
Pucci M, Micioni Di Bonaventura MV, Wille-Bille A, Fernández MS, Maccarrone M, Pautassi RM, Cifani C, D’Addario C. Environmental stressors and alcoholism development: Focus on molecular targets and their epigenetic regulation. Neurosci Biobehav Rev 2019; 106:165-181. [DOI: 10.1016/j.neubiorev.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/13/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023]
|
14
|
Newman EL, Leonard MZ, Arena DT, de Almeida RMM, Miczek KA. Social defeat stress and escalation of cocaine and alcohol consumption: Focus on CRF. Neurobiol Stress 2018; 9:151-165. [PMID: 30450381 PMCID: PMC6236516 DOI: 10.1016/j.ynstr.2018.09.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
Both the ostensibly aversive effects of unpredictable episodes of social stress and the intensely rewarding effects of drugs of abuse activate the mesocorticolimbic dopamine systems. Significant neuroadaptations in interacting stress and reward neurocircuitry may underlie the striking connection between stress and substance use disorders. In rodent models, recurring intermittent exposure to social defeat stress appears to produce a distinct profile of neuroadaptations that translates most readily to the repercussions of social stress in humans. In the present review, preclinical rodent models of social defeat stress and subsequent alcohol, cocaine or opioid consumption are discussed with regard to: (1) the temporal pattern of social defeat stress, (2) male and female protocols of social stress-escalated drug consumption, and (3) the neuroplastic effects of social stress, which may contribute to escalated drug-taking. Neuroadaptations in corticotropin-releasing factor (CRF) and CRF modulation of monoamines in the ventral tegmental area and the bed nucleus of the stria terminalis are highlighted as potential mechanisms underlying stress-escalated drug consumption. However, the specific mechanisms that drive CRF-mediated increases in dopamine require additional investigation as do the stress-induced neuroadaptations that may contribute to the development of compulsive patterns of drug-taking.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Dept., Tufts University, Medford, MA, 02155, USA
| | | | | | - Rosa M M de Almeida
- Institute of Psychology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Klaus A Miczek
- Psychology Dept., Tufts University, Medford, MA, 02155, USA.,Dept. of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, 02111, USA
| |
Collapse
|
15
|
Broccoli L, Uhrig S, von Jonquieres G, Schönig K, Bartsch D, Justice NJ, Spanagel R, Sommer W, Klugmann M, Hansson A. Targeted overexpression of CRH receptor subtype 1 in central amygdala neurons: effect on alcohol-seeking behavior. Psychopharmacology (Berl) 2018; 235:1821-1833. [PMID: 29700576 PMCID: PMC7454014 DOI: 10.1007/s00213-018-4908-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 04/12/2018] [Indexed: 12/14/2022]
Abstract
RATIONALE The corticotropin-releasing hormone (CRH) system is a key mediator of stress-induced responses in alcohol-seeking behavior. Recent research has identified the central nucleus of the amygdala (CeA), a brain region involved in the regulation of fear and stress-induced responses that is especially rich in CRH-positive neurons, as a key player in mediating excessive alcohol seeking. However, detailed characterization of the specific influences that local neuronal populations exert in mediating alcohol responses is hampered by current limitations in pharmacological and immunohistochemical tools for targeting CRH receptor subtype 1 (CRHR1). OBJECTIVE In this study, we investigated the effect of cell- and region-specific overexpression of CRHR1 in the CeA using a novel transgenic tool. METHODS Co-expression of CRHR1 in calcium-calmodulin-dependent kinase II (αCaMKII) neurons of the amygdala was demonstrated by double immunohistochemistry using a Crhr1-GFP reporter mouse line. A Cre-inducible Crhr1-expressing adeno-associated virus (AAV) was site-specifically injected into the CeA of αCaMKII-CreERT2 transgenic rats to analyze the role of CRHR1 in αCaMKII neurons on alcohol self-administration and reinstatement behavior. RESULTS Forty-eight percent of CRHR1-containing cells showed co-expression of αCaMKII in the CeA. AAV-mediated gene transfer in αCaMKII neurons induced a 24-fold increase of Crhr1 mRNA in the CeA which had no effect on locomotor activity, alcohol self-administration, or cue-induced reinstatement. However, rats overexpressing Crhr1 in the CeA increased responding in the stress-induced reinstatement task with yohimbine serving as a pharmacological stressor. CONCLUSION We demonstrate that CRHR1 overexpression in CeA-αCaMKII neurons is sufficient to mediate increased vulnerability to stress-triggered relapse into alcohol seeking.
Collapse
Affiliation(s)
- L. Broccoli
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - S. Uhrig
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - G. von Jonquieres
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia Sydney, NSW, Australia
| | - K. Schönig
- Dept. of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Square J5, 68159 Mannheim, Germany
| | - D. Bartsch
- Dept. of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Square J5, 68159 Mannheim, Germany
| | - N. J. Justice
- Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, Texas 77030, USA
| | - R. Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - W.H. Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany,Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| | - M. Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia Sydney, NSW, Australia
| | - A.C. Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany,To whom correspondence should be addressed: Anita C. Hansson, PhD, Institute of Psychopharmacology, Central Institute for Mental Health, University of Heidelberg, Medical Faculty Mannheim, Square J5, D-68159 Mannheim, Germany, Phone: +49 621 1703 6293, Fax: +49 621 1703 6255,
| |
Collapse
|
16
|
Newman EL, Albrechet-Souza L, Andrew PM, Auld JG, Burk KC, Hwa LS, Zhang EY, DeBold JF, Miczek KA. Persistent escalation of alcohol consumption by mice exposed to brief episodes of social defeat stress: suppression by CRF-R1 antagonism. Psychopharmacology (Berl) 2018; 235:1807-1820. [PMID: 29696309 PMCID: PMC6168197 DOI: 10.1007/s00213-018-4905-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
RATIONALE Episodic bouts of social stress can precede the initiation, escalation, or relapse to disordered alcohol intake. Social stress may engender neuroadaptations in the hypothalamic-pituitary-adrenal (HPA) axis and in extrahypothalamic stress circuitry to promote the escalation of alcohol intake. OBJECTIVES We aimed to (1) confirm a pattern of escalated drinking in socially defeated mice and to (2) test drugs that target distinct aspects of the HPA axis and extrahypothalamic neural substrates for their effectiveness in reducing murine, stress-escalated drinking. METHODS Male C57BL/6J (B6) mice were socially defeated by resident Swiss-derived males for ten consecutive days receiving 30 bites/day. Ten days after the final defeat, cohorts of B6 mice received continuous or intermittent access to 20% EtOH (w/v) and water. After 4 weeks of drinking, mice were injected with weekly, systemic doses of the CRF-R1 antagonist, CP376395; the glucocorticoid receptor antagonist, mifepristone; the 11-beta-hydroxylase inhibitor, metyrapone; or the 5-alpha-reductase inhibitor, finasteride. RESULTS Prior to drug treatments, defeated mice reliably consumed more EtOH than non-defeated controls, and mice given alcohol intermittently consumed more EtOH than those with continuous access. CP376395 (17-30 mg/kg) reduced continuous, but not intermittent EtOH intake (g/kg) in socially defeated mice. Mifepristone (100 mg/kg), however, increased drinking by defeated mice with intermittent access to alcohol while reducing drinking during continuous access. When administered finasteride (100 mg/kg) or metyrapone (50 mg/kg), all mice reduced their EtOH intake while increasing their water consumption. CONCLUSIONS Mice with a history of episodic social defeat stress were selectively sensitive to the effects of CRF-R1 antagonism, suggesting that CRF-R1 may be a potential target for treating alcohol use disorders in individuals who escalate their drinking after exposure to repeated bouts of psychosocial stress. Future studies will clarify how social defeat stress may alter the expression of extrahypothalamic CRF-R1 and glucocorticoid receptors.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Lucas Albrechet-Souza
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Peter M Andrew
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - John G Auld
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Kelly C Burk
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Lara S Hwa
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Eric Y Zhang
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Joseph F DeBold
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Klaus A Miczek
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA.
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, 02111, USA.
| |
Collapse
|
17
|
Nelson BS, Sequeira MK, Schank JR. Bidirectional relationship between alcohol intake and sensitivity to social defeat: association with Tacr1 and Avp expression. Addict Biol 2018; 23:142-153. [PMID: 28150369 DOI: 10.1111/adb.12494] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/01/2017] [Accepted: 01/04/2017] [Indexed: 01/17/2023]
Abstract
While epidemiological studies show that alcohol abuse is often co-morbid with affective disorders, the causal direction of this association is unclear. We examined this relationship using mouse models including social defeat stress (SDS), social interaction (SI) and voluntary alcohol consumption. C57BL6/J mice exposed to SDS segregate into two subpopulations, those that express depressive-like phenotypes ('susceptible') and those that do not ('resilient'). First, we stratified SDS-exposed mice and measured their voluntary alcohol consumption. Next, we determined whether SI behavior in alcohol-naïve mice could predict alcohol intake. Finally, we assessed the effect of binge-like alcohol exposure on sensitivity to SDS. We quantified Tacr1 (neurokinin-1 receptor gene) and Avp (vasopressin peptide gene) mRNA in brain regions involved in depression, addiction and social behavior. We found that susceptible mice consumed more alcohol compared with resilient mice, suggesting that depression-like phenotypes associate with increased alcohol intake. Interestingly, we observed a negative correlation between SI and alcohol intake in stress- and alcohol-naïve mice, suggesting that individual differences in SI associate with alcohol preference. Finally, alcohol pre-treatment increased sensitivity to SDS, indicating that alcohol exposure alters sensitivity to social stress. Quantification of mRNA revealed that increased expression of Tacr1 and Avp generally associated with decreased SI and increased alcohol intake. C57BL6/J mice are an inbred strain; thus, it is likely that individual differences in behavior and gene expression are driven by epigenetic factors. Collectively, these results support a bidirectional relationship between alcohol exposure and susceptibility to stress that is associated with variations in neuropeptide expression.
Collapse
Affiliation(s)
- Britta S. Nelson
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| | - Michelle K. Sequeira
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| | - Jesse R. Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine; University of Georgia; GA USA
| |
Collapse
|
18
|
Pomrenze MB, Fetterly TL, Winder DG, Messing RO. The Corticotropin Releasing Factor Receptor 1 in Alcohol Use Disorder: Still a Valid Drug Target? Alcohol Clin Exp Res 2017; 41:1986-1999. [PMID: 28940382 PMCID: PMC5711524 DOI: 10.1111/acer.13507] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/15/2017] [Indexed: 01/20/2023]
Abstract
Corticotropin releasing factor (CRF) is a neuropeptide that plays a key role in behavioral and physiological responses to stress. A large body of animal literature implicates CRF acting at type 1 CRF receptors (CRFR1) in consumption by alcohol-dependent subjects, stress-induced reinstatement of alcohol seeking, and possibly binge alcohol consumption. These studies have encouraged recent pilot studies of CRFR1 antagonists in humans with alcohol use disorder (AUD). It was a great disappointment to many in the field that these studies failed to show an effect of these compounds on stress-induced alcohol craving. Here, we examine these studies to explore potential limitations and discuss preclinical and human literature to ask whether CRFR1 is still a valid drug target to pursue for the treatment of AUD.
Collapse
Affiliation(s)
| | - Tracy L. Fetterly
- Department of Molecular Physiology & Biophysics, Vanderbilt
University, Nashville, TN
- Vanderbilt Neuroscience Graduate Program, Vanderbilt University,
Nashville, TN
| | - Danny G. Winder
- Department of Molecular Physiology & Biophysics, Vanderbilt
University, Nashville, TN
- Vanderbilt Center for Addiction Research, Vanderbilt University,
Nashville, TN
- Vanderbilt Neuroscience Graduate Program, Vanderbilt University,
Nashville, TN
| | - Robert O. Messing
- Institute for Neuroscience, University of Texas at Austin, Austin,
TX
- Departments of Neuroscience and Neurology, University of Texas at
Austin, Austin, TX
- Waggoner Center for Alcohol and Addiction Research, University of
Texas at Austin, Austin, TX
| |
Collapse
|
19
|
Albrechet-Souza L, Viola TW, Grassi-Oliveira R, Miczek KA, de Almeida RMM. Corticotropin Releasing Factor in the Bed Nucleus of the Stria Terminalis in Socially Defeated and Non-stressed Mice with a History of Chronic Alcohol Intake. Front Pharmacol 2017; 8:762. [PMID: 29118713 PMCID: PMC5660971 DOI: 10.3389/fphar.2017.00762] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/10/2017] [Indexed: 11/21/2022] Open
Abstract
Stress exposure has been identified as one risk factor for alcohol abuse that may facilitate the transition from social or regulated use to the development of alcohol dependence. Preclinical studies have shown that dysregulation of the corticotropin releasing factor (CRF) neurotransmission has been implicated in stress-related psychopathologies such as depression and anxiety, and may affect alcohol consumption. The bed nucleus of the stria terminalis (BNST) contains CRF-producing neurons which seem to be sensitive to stress. In this study, adult male C57BL/6 mice previously defeated in resident-intruder confrontations were evaluated in the elevated plus-maze and tail suspension test. Mice were also tested for sweet solution intake before and after social stress. After having had continuous access to ethanol (20% weight/volume) for 4 weeks, control and stressed mice had CRF type 1 (CRFR1) or type 2 (CRFR2) receptor antagonists infused into the BNST and then had access to ethanol for 24 h. In separate cohorts of control and stressed mice, we assessed mRNA levels of BNST CRF, CRFR1 and CRFR2. Stressed mice increased their intake of sweet solution after ten sessions of social defeat and showed reduced activity in the open arms of the elevated plus-maze. When tested for ethanol consumption, stressed mice persistently drank significantly more than controls during the 4 weeks of access. Also, social stress induced higher BNST CRF mRNA levels. The selective blockade of BNST CRFR1 with CP376,395 effectively reduced alcohol drinking in non-stressed mice, whereas the selective CRFR2 antagonist astressin2B produced a dose-dependent increase in ethanol consumption in both non-stressed controls and stressed mice. The 10-day episodic defeat stress used here elicited anxiety- but not depressive-like behaviors, and promoted an increase in ethanol drinking. CRF-CRFR1 signaling in the BNST seems to underlie ethanol intake in non-stressed mice, whereas CRFR2 modulates alcohol consumption in both socially defeated and non-stressed mice with a history of chronic intake.
Collapse
Affiliation(s)
- Lucas Albrechet-Souza
- Institute of Psychology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thiago W Viola
- Developmental Cognitive Neuroscience Lab (DCNL) and Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL) and Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Klaus A Miczek
- Departments of Psychology and Neuroscience, Tufts University, Medford, MA, United States
| | - Rosa M M de Almeida
- Institute of Psychology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
20
|
Tanaka M, Tomimatsu Y, Sakimura K, Ootani Y, Sako Y, Kojima T, Aso K, Yano T, Hirai K. Characterization of CRF 1 receptor antagonists with differential peripheral vs central actions in CRF challenge in rats. Peptides 2017; 95:40-50. [PMID: 28689880 DOI: 10.1016/j.peptides.2017.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 06/20/2017] [Accepted: 06/26/2017] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate peripheral and central roles of corticotropin-releasing factor (CRF) in endocrinological and behavioral changes. Plasma adrenocorticotropin (ACTH) concentration was measured as an activity of hypothalamic-pituitary-adrenal (HPA) axis. As behavioral changes, locomotion and anxiety behavior were measured after CRF challenge intravenously (i.v.) for the peripheral administration or intracerebroventricularly (i.c.v.) for the central administration. Plasma ACTH concentration was significantly increased by both administration routes of CRF; however, hyperlocomotion and anxiety behavior were induced only by the i.c.v. administration. In the drug discovery of CRF1 receptor antagonists, we identified two types of compounds, Compound A and Compound B, which antagonized peripheral CRF-induced HPA axis activation to the same extent, but showed different effects on the central CRF signal. These had similar in vitro CRF1 receptor binding affinities (15 and 10nM) and functional activities in reporter gene assay (15 and 9.5nM). In the ex vivo binding assays using tissues of the pituitary, oral treatment with Compound A and Compound B at 10mg/kg inhibited [125I]-CRF binding, whereas in the assay using tissues of the frontal cortex, treatment of Compound A but not Compound B inhibited [125I]-CRF binding, indicating that only Compound A inhibited central [125I]-CRF binding. In the peripheral CRF challenge, increase in plasma ACTH concentration was significantly suppressed by both Compound A and Compound B. In contrast, Compound A inhibited the increase in locomotion induced by the central CRF challenge while Compound B did not. Compound A also reduced central CRF challenge-induced anxiety behavior and c-fos immunoreactivity in the cortex and the hypothalamic paraventricular nucleus. These results indicate that the central CRF signal, rather than the peripheral CRF signal would be related to anxiety and other behavioral changes, and CRF1 receptor antagonism in the central nervous system may be critical for identifying drug candidates for anxiety and mood disorders.
Collapse
Affiliation(s)
- Maiko Tanaka
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yoshiro Tomimatsu
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Katsuya Sakimura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yoshikazu Ootani
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuu Sako
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takuto Kojima
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazuyoshi Aso
- Research Alliance Group, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takahiko Yano
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Keisuke Hirai
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
21
|
Contribution of Urocortin to the Development of Excessive Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:275-291. [PMID: 29056154 DOI: 10.1016/bs.irn.2017.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The corticotropin-releasing factor (CRF) system plays a role in alcohol consumption, and its dysregulation can contribute to alcohol use disorder. This system includes four peptide ligands: CRF, urocortin (Ucn)1, Ucn2, and Ucn3. Historically, attention focused on CRF, however, Ucn1 also plays a critical role in excessive alcohol use. This review covers evidence for this contribution and contrasts the role of Ucn1 with CRF. While CRF can promote binge consumption, this regulation occurs through generalized mechanisms that are not specific for alcohol. In contrast, inhibition of Ucn1 action specifically blunts escalation of alcohol drinking. Lesions, genetic knockout, and RNA interference experiments indicate that the centrally projecting Edinger-Westphal nucleus is the neuroanatomical source of Ucn1 critical for alcohol drinking. We propose that the contributions of Ucn1 to excessive drinking likely occur through enhancing rewarding properties of alcohol and symptoms of alcohol withdrawal, whereas CRF drives dependence-induced drinking at later stages of alcohol use. The transition from occasional binge drinking to dependence intricately depends on CRF system plasticity and coordination of CRF and Ucn1.
Collapse
|
22
|
Synergistic blockade of alcohol escalation drinking in mice by a combination of novel kappa opioid receptor agonist Mesyl Salvinorin B and naltrexone. Brain Res 2017; 1662:75-86. [PMID: 28263712 DOI: 10.1016/j.brainres.2017.02.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 02/23/2017] [Accepted: 02/27/2017] [Indexed: 12/29/2022]
Abstract
Mesyl Salvinorin B (MSB) is a potent selective kappa opioid receptor (KOP-r) agonist that has potential for development as an anti-psychostimulant agent with fewer side-effects (e.g., sedation, depression and dysphoria) than classic KOP-r agonists. However, no such study has been done on alcohol. We investigated whether MSB alone or in combination with naltrexone (mu-opioid receptor antagonist) altered voluntary alcohol drinking in both male and female mice. Mice, subjected to 3weeks of chronic escalation drinking (CED) in a two-bottle choice paradigm with 24-h access every other day, developed rapid escalation of alcohol intake and high preference. We found that single, acute administration of MSB dose-dependently reduced alcohol intake and preference in mice after 3-week CED. The effect was specific to alcohol, as shown by the lack of any effect of MSB on sucrose or saccharin intake. We also used the drinking-in-the-dark (DID) model with limited access (4h/day) to evaluate the pharmacological effect of MSB after 3weeks of DID. However, MSB had no effect on alcohol drinking after 3-week DID. Upon investigation of potential synergistic effects between naltrexone and MSB, we found that acute administration of a combination of MSB and naltrexone reduced alcohol intake profoundly after 3-week CED at doses lower than those individual effective doses. Repeated administrations of this combination showed less tolerance development than repeated MSB alone. Our study suggests that the novel KOP-r agonist MSB both alone and in combination with naltrexone shows potential in alcoholism treatment models.
Collapse
|
23
|
Contoreggi C, Chrousos GP, Mascio MD. Chronic distress and the vulnerable host: a new target for HIV treatment and prevention? NEUROBEHAVIORAL HIV MEDICINE 2016; 7:53-75. [PMID: 34295195 PMCID: PMC8293862 DOI: 10.2147/nbhiv.s86309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pathologic stress (distress) disturbs immune, cardiovascular, metabolic, and behavioral homeostasis. Individuals living with HIV and those at risk are vulnerable to stress disorders. Corticotropin-releasing hormone (CRH) is critical in neuroendocrine immune regulation. CRH, a neuropeptide, is distributed in the central and peripheral nervous systems and acts principally on CRH receptor type 1 (CRHR1). CRH in the brain modulates neuropsychiatric disorders. CRH and stress modulation of immunity is two-pronged; there is a direct action on hypothalamic-pituitary-adrenal secretion of glucocorticoids and through immune organ sympathetic innervation. CRH is a central and systemic proinflammatory cytokine. Glucocorticoids and their receptors have gene regulatory actions on viral replication and cause central and systemic immune suppression. CRH and stress activation contributes to central nervous system (CNS) viral entry important in HIV-associated neurocognitive disorders and HIV-associated dementia. CNS CRH overproduction short-circuits reward, executive, and emotional control, leading to addiction, cognitive impairment, and psychiatric comorbidity. CRHR1 is an important therapeutic target for medication development. CRHR1 antagonist clinical trials have focused on psychiatric disorders with little attention paid to neuroendocrine immune disorders. Studies of those with HIV and those at risk show that concurrent stress-related disorders contribute to higher morbidity and mortality; stress-related conditions, addiction, immune dysfunction, and comorbid psychiatric illness all increase HIV transmission. Neuropsychiatric disease, chronic inflammation, and substance abuse are endemic, and chronic distress is a pathologic factor. It is being understood that stress and CRH are fundamental to neuroendocrine immunity; therapeutic interventions with existing and novel agents hold promise for restoring homeostasis, reducing morbidity and mortality for those with HIV and possibly reducing future disease transmission.
Collapse
Affiliation(s)
- Carlo Contoreggi
- Intramural Research Program (IRP), National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - George P Chrousos
- Department of Pediatrics, Aghia Sophia Children’s Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Michele Di Mascio
- AIDS Imaging Research Section, Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
24
|
Gilpin NW, Weiner JL. Neurobiology of comorbid post-traumatic stress disorder and alcohol-use disorder. GENES BRAIN AND BEHAVIOR 2016; 16:15-43. [PMID: 27749004 DOI: 10.1111/gbb.12349] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/03/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
Post-traumatic stress disorder (PTSD) and alcohol-use disorder (AUD) are highly comorbid in humans. Although we have some understanding of the structural and functional brain changes that define each of these disorders, and how those changes contribute to the behavioral symptoms that define them, little is known about the neurobiology of comorbid PTSD and AUD, which may be due in part to a scarcity of adequate animal models for examining this research question. The goal of this review is to summarize the current state-of-the-science on comorbid PTSD and AUD. We summarize epidemiological data documenting the prevalence of this comorbidity, review what is known about the potential neurobiological basis for the frequent co-occurrence of PTSD and AUD and discuss successes and failures of past and current treatment strategies. We also review animal models that aim to examine comorbid PTSD and AUD, highlighting where the models parallel the human condition, and we discuss the strengths and weaknesses of each model. We conclude by discussing key gaps in our knowledge and strategies for addressing them: in particular, we (1) highlight the need for better animal models of the comorbid condition and better clinical trial design, (2) emphasize the need for examination of subpopulation effects and individual differences and (3) urge cross-talk between basic and clinical researchers that is reflected in collaborative work with forward and reverse translational impact.
Collapse
Affiliation(s)
- N W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA.,Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - J L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
25
|
Ng E, Browne CJ, Samsom JN, Wong AHC. Depression and substance use comorbidity: What we have learned from animal studies. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:456-474. [PMID: 27315335 DOI: 10.1080/00952990.2016.1183020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Depression and substance use disorders are often comorbid, but the reasons for this are unclear. In human studies, it is difficult to determine how one disorder may affect predisposition to the other and what the underlying mechanisms might be. Instead, animal studies allow experimental induction of behaviors relevant to depression and drug-taking, and permit direct interrogation of changes to neural circuits and molecular pathways. While this field is still new, here we review animal studies that investigate whether depression-like states increase vulnerability to drug-taking behaviors. Since chronic psychosocial stress can precipitate or predispose to depression in humans, we review studies that use psychosocial stressors to produce depression-like phenotypes in animals. Specifically, we describe how postweaning isolation stress, repeated social defeat stress, and chronic mild (or unpredictable) stress affect behaviors relevant to substance abuse, especially operant self-administration. Potential brain changes mediating these effects are also discussed where available, with an emphasis on mesocorticolimbic dopamine circuits. Postweaning isolation stress and repeated social defeat generally increase acquisition or maintenance of drug self-administration, and alter dopamine sensitivity in various brain regions. However, the effects of chronic mild stress on drug-taking have been much less studied. Future studies should consider standardizing stress-induction protocols, including female subjects, and using multi-hit models (e.g. genetic vulnerabilities and environmental stress).
Collapse
Affiliation(s)
- Enoch Ng
- a Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada.,b Institute of Medical Science, University of Toronto , Toronto , Canada
| | - Caleb J Browne
- c Department of Psychology , University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada
| | - James N Samsom
- d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada
| | - Albert H C Wong
- b Institute of Medical Science, University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada.,f Department of Psychiatry , University of Toronto , Toronto , Canada
| |
Collapse
|
26
|
Morisot N, Contarino A. The CRF 1 and the CRF 2 receptor mediate recognition memory deficits and vulnerability induced by opiate withdrawal. Neuropharmacology 2016; 105:500-507. [DOI: 10.1016/j.neuropharm.2016.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 11/17/2022]
|
27
|
Effect of different stressors on voluntary ethanol intake in ethanol-dependent and nondependent C57BL/6J mice. Alcohol 2016; 51:17-23. [PMID: 26992696 DOI: 10.1016/j.alcohol.2015.11.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022]
Abstract
Several animal models have evaluated the effect of stress on voluntary ethanol intake with mixed results. The experiments reported here examined the effects of different stressors on voluntary ethanol consumption in dependent and nondependent adult male C57BL/6J mice. In Experiment 1, restraint, forced swim, and social defeat stress procedures all tended to reduce ethanol intake in nondependent mice regardless of whether the stress experience occurred 1 h or 4 h prior to ethanol access. The reduction in ethanol consumption was most robust following restraint stress. Experiment 2 examined the effects of forced swim stress and social defeat stress on drinking in a dependence model that involved repeated cycles of chronic intermittent ethanol (CIE) exposure. Repeated exposure to forced swim stress prior to intervening test drinking periods that followed repeated cycles of CIE exposure further increased ethanol consumption in CIE-exposed mice while not altering intake in nondependent mice. In contrast, repeated exposure to the social defeat stressor in a similar manner reduced ethanol consumption in CIE-exposed mice while not altering drinking in nondependent mice. Results from Experiment 3 confirmed this selective effect of forced swim stress increasing ethanol consumption in mice with a history of CIE exposure, and also demonstrated that enhanced drinking is only observed when the forced swim stressor is administered during each test drinking week, but not if it is applied only during the final test week. Collectively, these studies point to a unique interaction between repeated stress experience and CIE exposure, and also suggest that such an effect depends on the nature of the stressor. Future studies will need to further explore the generalizability of these results, as well as mechanisms underlying the ability of forced swim stress to selectively further enhance ethanol consumption in dependent (CIE-exposed) mice but not alter intake in nondependent animals.
Collapse
|
28
|
Anderson RI, Lopez MF, Becker HC. Stress-Induced Enhancement of Ethanol Intake in C57BL/6J Mice with a History of Chronic Ethanol Exposure: Involvement of Kappa Opioid Receptors. Front Cell Neurosci 2016; 10:45. [PMID: 26941607 PMCID: PMC4763044 DOI: 10.3389/fncel.2016.00045] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/08/2016] [Indexed: 12/22/2022] Open
Abstract
Our laboratory has previously demonstrated that daily forced swim stress (FSS) prior to ethanol drinking sessions facilitates enhanced ethanol consumption in mice with a history of chronic intermittent ethanol (CIE) vapor exposure without altering ethanol intake in air-exposed controls. Because both stress and chronic ethanol exposure have been shown to activate the dynorphin/kappa opioid receptor (KOR) system, the present study was designed to explore a potential role for KORs in modulating stress effects on ethanol consumption in the CIE model of dependence and relapse drinking. After stable baseline ethanol intake was established in adult male C57BL/6J mice, subjects received chronic intermittent exposure (16 h/day × 4 days/week) to ethanol vapor (CIE group) or air (CTL group). Weekly cycles of inhalation exposure were alternated with 5-day limited access drinking tests (1 h access to 15% ethanol). Experiment 1 compared effects of daily FSS and KOR activation on ethanol consumption. CIE and CTL mice were either exposed to FSS (10 min), the KOR agonist U50,488 (5 mg/kg), or a vehicle injection (non-stressed condition) prior to each daily drinking session during test weeks. FSS selectively increased drinking in CIE mice. U50,488 mimicked this effect in CIE mice, but also increased drinking in CTL mice. Experiment 2 assessed effects of KOR blockade on stress-induced drinking in CIE and CTL mice. Stressed and non-stressed mice were administered the short-acting KOR antagonist LY2444296 (0 or 5 mg/kg) 30 min prior to each drinking session during test weeks. FSS selectively increased ethanol consumption in CIE mice, an effect that was abolished by LY2444296 pretreatment. In Experiment 3, CIE and CTL mice were administered one of four doses of U50,488 (0, 1.25, 2.5, 5.0 mg/kg) 1 h prior to each daily drinking test (in lieu of FSS). All doses of U50,488 increased ethanol consumption in both CIE and CTL mice. The U50,488-induced increase in drinking was blocked by LY2444296. Our results demonstrate that the KOR system contributes to the stress enhancement of ethanol intake in mice with a history of chronic ethanol exposure.
Collapse
Affiliation(s)
- Rachel I Anderson
- Medical University of South CarolinaCharleston, SC, USA; Charleston Alcohol Research CenterCharleston, SC, USA
| | - Marcelo F Lopez
- Medical University of South CarolinaCharleston, SC, USA; Charleston Alcohol Research CenterCharleston, SC, USA
| | - Howard C Becker
- Medical University of South CarolinaCharleston, SC, USA; Charleston Alcohol Research CenterCharleston, SC, USA; Ralph H. Johnson Veterans Administration Medical CenterCharleston, SC, USA
| |
Collapse
|
29
|
Hwa LS, Holly EN, DeBold JF, Miczek KA. Social stress-escalated intermittent alcohol drinking: modulation by CRF-R1 in the ventral tegmental area and accumbal dopamine in mice. Psychopharmacology (Berl) 2016; 233:681-90. [PMID: 26576941 PMCID: PMC4729595 DOI: 10.1007/s00213-015-4144-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 10/29/2015] [Indexed: 12/22/2022]
Abstract
RATIONALE Excessive alcohol (EtOH) drinking is difficult to model in animals despite the extensive human literature demonstrating that stress increases EtOH consumption. OBJECTIVE The current experiments show escalations in voluntary EtOH drinking caused by a history of social defeat stress and intermittent access to EtOH in C57BL/6J mice compared to non-stressed mice given intermittent EtOH or continuous EtOH. To explore a mechanistic link between stress and drinking, we studied the role of corticotropin-releasing factor type-1 receptors (CRF-R1) in the dopamine-rich ventral tegmental area (VTA). RESULTS Intra-VTA infusions of a CRF-R1 antagonist, CP376395, infused into the VTA dose-dependently and selectively reduced intermittent EtOH intake in stressed and non-stressed mice, but not in mice given continuous EtOH. In contrast, intra-VTA infusions of the CRF-R2 antagonist astressin2B non-specifically suppressed both EtOH and H2O drinking in the stressed group without effects in the non-stressed mice. Using in vivo microdialysis in the nucleus accumbens (NAc) shell, we observed that stressed mice drinking EtOH intermittently had elevated levels of tonic dopamine concentrations compared to non-stressed drinking mice. Also, VTA CP376395 potentiated dopamine output to the NAc only in the stressed group causing further elevations of dopamine post-infusion. CONCLUSIONS These findings illustrate a role for extrahypothalamic CRF-R1 as especially important for stress-escalated EtOH drinking beyond schedule-escalated EtOH drinking. CRF-R1 may be a mechanism for balancing the dysregulation of stress and reward in alcohol use disorders.
Collapse
Affiliation(s)
- Lara S Hwa
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Elizabeth N Holly
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Joseph F DeBold
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Klaus A Miczek
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA.
- Neuroscience Department, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
30
|
Heilig M, Sommer WH, Spanagel R. The Need for Treatment Responsive Translational Biomarkers in Alcoholism Research. Curr Top Behav Neurosci 2016; 28:151-171. [PMID: 27240677 DOI: 10.1007/7854_2015_5006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Over the past two decades, major advances have been made in the basic neuroscience of alcohol addiction. However, few of these have been translated into clinically useful treatments, which remain limited. In the past decade, psychiatric drug development in general has been stalled, with many preclinically validated mechanisms failing in clinical development. Despite the existence of appealing preclinical models in the area of addictive disorders, drug development for these conditions has been impacted by the exodus of major pharma from psychiatric neuroscience. Here, we discuss translational biomarker strategies that may help turn this tide. Following an approach patterned on an endophenotype approach to complex behavioral traits, we hypothesize that relatively simple biological measures should be sought that can be obtained both in experimental animals and in humans, and that may be responsive to alcoholism medications. These biomarkers have to be tailored to the specific mechanism targeted by candidate medications and may in fact also help identify biologically more homogeneous subpopulations of patients. We introduce as examples alcohol-induced dopamine (DA) release, measures of central glutamate levels, and network connectivity, and discuss our experience to date with these biomarker strategies.
Collapse
Affiliation(s)
- Markus Heilig
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, SE-58183, Linköping, Sweden.
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Mayfield J, Arends MA, Harris RA, Blednov YA. Genes and Alcohol Consumption: Studies with Mutant Mice. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:293-355. [PMID: 27055617 PMCID: PMC5302130 DOI: 10.1016/bs.irn.2016.02.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter, we review the effects of global null mutant and overexpressing transgenic mouse lines on voluntary self-administration of alcohol. We examine approximately 200 publications pertaining to the effects of 155 mouse genes on alcohol consumption in different drinking models. The targeted genes vary in function and include neurotransmitter, ion channel, neuroimmune, and neuropeptide signaling systems. The alcohol self-administration models include operant conditioning, two- and four-bottle choice continuous and intermittent access, drinking in the dark limited access, chronic intermittent ethanol, and scheduled high alcohol consumption tests. Comparisons of different drinking models using the same mutant mice are potentially the most informative, and we will highlight those examples. More mutants have been tested for continuous two-bottle choice consumption than any other test; of the 137 mouse genes examined using this model, 97 (72%) altered drinking in at least one sex. Overall, the effects of genetic manipulations on alcohol drinking often depend on the sex of the mice, alcohol concentration and time of access, genetic background, as well as the drinking test.
Collapse
Affiliation(s)
- J Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - M A Arends
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, United States
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
32
|
Shirahase T, Aoki M, Watanabe R, Watanabe Y, Tanaka M. Increased alcohol consumption in relaxin-3 deficient male mice. Neurosci Lett 2015; 612:155-160. [PMID: 26687275 DOI: 10.1016/j.neulet.2015.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/03/2015] [Accepted: 12/09/2015] [Indexed: 11/29/2022]
Abstract
Relaxin-3 is a neuropeptide expressed in the brainstem, and predominantly localized in the gray matter of the midline dorsal pons termed the nucleus incertus. Relaxin-3-expressing neurons densely project axons rostrally to various forebrain regions including the septum, hippocampus, and lateral hypothalamus. Several relaxin-3 functions have been reported including food intake, stress responses, neuroendocrine function, emotion, and spatial memory. In addition, recently relaxin-3 and its receptor, RXFP3, were shown to regulate alcohol intake using an RXFP3 antagonist and RXFP3 gene knockout mice. In the present study, we investigated alcohol consumption in relaxin-3 knockout mice, and found that male but not female mice significantly drank more alcohol than wild-type mice in the two-bottle choice test. However, after chronic alcohol vapor exposure, wild-type and mutant mice did not show this difference in alcohol intake, although both genotypes exhibited increased alcohol consumption compared with non-alcohol-exposed control mice. There was no genotype difference in sucrose or quinine preference. These results suggest that the relaxin-3 neuronal system modestly affects alcohol preference and consumption.
Collapse
Affiliation(s)
- Takahira Shirahase
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan; Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Miku Aoki
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan; Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Ryuji Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Yoshihisa Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Masaki Tanaka
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
33
|
Miczek KA, DeBold JF, Hwa LS, Newman EL, de Almeida RMM. Alcohol and violence: neuropeptidergic modulation of monoamine systems. Ann N Y Acad Sci 2015; 1349:96-118. [PMID: 26285061 DOI: 10.1111/nyas.12862] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurobiological processes underlying the epidemiologically established link between alcohol and several types of social, aggressive, and violent behavior remain poorly understood. Acute low doses of alcohol, as well as withdrawal from long-term alcohol use, may lead to escalated aggressive behavior in a subset of individuals. An urgent task will be to disentangle the host of interacting genetic and environmental risk factors in individuals who are predisposed to engage in escalated aggressive behavior. The modulation of 5-hydroxytryptamine impulse flow by gamma-aminobutyric acid (GABA) and glutamate, acting via distinct ionotropic and metabotropic receptor subtypes in the dorsal raphe nucleus during alcohol consumption, is of critical significance in the suppression and escalation of aggressive behavior. In anticipation and reaction to aggressive behavior, neuropeptides such as corticotropin-releasing factor, neuropeptide Y, opioid peptides, and vasopressin interact with monoamines, GABA, and glutamate to attenuate and amplify aggressive behavior in alcohol-consuming individuals. These neuromodulators represent novel molecular targets for intervention that await clinical validation. Intermittent episodes of brief social defeat during aggressive confrontations are sufficient to cause long-lasting neuroadaptations that can lead to the escalation of alcohol consumption.
Collapse
Affiliation(s)
- Klaus A Miczek
- Departments of Pharmacology, Psychiatry, and Neuroscience, Tufts University, Boston, Massachusetts.,Department of Psychology, Tufts University, Medford, Massachusetts
| | - Joseph F DeBold
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Lara S Hwa
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Emily L Newman
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Rosa M M de Almeida
- Department of Psychology, LPNeC, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
34
|
Reduction of ethanol intake by corticotropin-releasing factor receptor-1 antagonist in "heavy-drinking" mice in a free-choice paradigm. Psychopharmacology (Berl) 2015; 232:2731-9. [PMID: 25797192 DOI: 10.1007/s00213-015-3909-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 02/25/2015] [Indexed: 12/16/2022]
Abstract
RATIONALE We hypothesized that the corticotropin-releasing factor (CRF) system is hyperresponsive in animals with high ethanol intake, which exhibits a reduction of ethanol intake when administered with a CRF1 receptor antagonist. METHODS Outbred Swiss mice were subjected to a long-term, three-bottle, free-choice paradigm (5 and 10 % [v/v] ethanol and water) that consisted of four phases: acquisition (AC; 10 weeks), withdrawal (W; 2 weeks), reexposure (RE; 2 weeks), and quinine-adulteration (AD; 2 weeks). Based on individual ethanol intake, the mice were classified into three groups: A group, preference for ethanol and persistently high consumption during AD phase; B group, preference for ethanol and a reduction of ethanol intake in the AD phase; and C group; preference for water during all phases. A control group only had access to water. CRF1 receptor messenger RNA (mRNA) levels in the amygdala and the effect of the CRF1 receptor antagonist CP-154,526 on ethanol and water intake in the subgroups were studied. RESULTS CRF1 transcript levels were higher in the B group than in the control group. The highest dose of CP-154,526 reduced ethanol intake and preference, with no changes in water consumption, in the A group compared with vehicle. The B group exhibited a reduction of both ethanol and water intake, with no changes in preference. The C group exhibited no changes in response to the CRF1 antagonist. CONCLUSIONS CRF1 receptors appear to be involved in ethanol consumption in mice with high ethanol consumption, and CRF system-mediated neuroadaptations depend on drinking profiles.
Collapse
|
35
|
Jimenez VA, Helms CM, Cornea A, Meshul CK, Grant KA. An ultrastructural analysis of the effects of ethanol self-administration on the hypothalamic paraventricular nucleus in rhesus macaques. Front Cell Neurosci 2015; 9:260. [PMID: 26236193 PMCID: PMC4500925 DOI: 10.3389/fncel.2015.00260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/23/2015] [Indexed: 01/29/2023] Open
Abstract
A bidirectional relationship between stress and ethanol exists whereby stressful events are comorbid with problematic ethanol use and prolonged ethanol exposure results in adaptations of the physiological stress response. Endocrine response to stress is initiated in the hypothalamic paraventricular nucleus (PVN) with the synthesis and release of corticotropin-releasing hormone (CRH) and arginine-vasopressin (AVP). Alterations in CRH and AVP following long-term ethanol exposure in rodents is well demonstrated, however little is known about the response to ethanol in primates or the mechanisms of adaptation. We hypothesized that long-term ethanol self-administration in nonhuman primates would lead to ultrastructural changes in the PVN underlying adaptation to chronic ethanol. Double-label immunogold electron microscopy (EM) was used to measure presynaptic gamma-aminobutyric acid (GABA) and glutamate density within synaptic terminals contacting CRH- and AVP-immunoreactive dendrites. Additionally, pituitary-adrenal hormones (ACTH, cortisol, DHEA-s and aldosterone) under two conditions (low and mild stress) were compared before and after self-administration. All hormones were elevated in response to the mild stressor independent of ethanol consumption. The presynaptic glutamate density in recurrent (i.e., intra-hypothalamic) CRH terminals was highly related to ethanol intake, and may be a permissive factor in increased drinking due to stress. Conversely, glutamate density within recurrent AVP terminals showed a trend-level increase following ethanol, but was not related to average daily consumption. Glutamate density in non-recurrent AVP terminals was related to aldosterone under the low stress condition while GABAergic density in this terminal population was related to water consumption. The results reveal distinct populations of presynaptic terminals whose glutamatergic or GABAergic density were uniquely related to water and ethanol consumption and circulating hormones.
Collapse
Affiliation(s)
- Vanessa A Jimenez
- Behavioral Neuroscience, Oregon Health and Science University Portland, OR, USA ; Division of Neuroscience, Oregon National Primate Research Center Beaverton, OR, USA
| | - Christa M Helms
- Division of Neuroscience, Oregon National Primate Research Center Beaverton, OR, USA
| | - Anda Cornea
- Division of Neuroscience, Oregon National Primate Research Center Beaverton, OR, USA
| | - Charles K Meshul
- Behavioral Neuroscience, Oregon Health and Science University Portland, OR, USA ; Research Services, Veterans Affairs Medical Center Portland, OR, USA
| | - Kathleen A Grant
- Behavioral Neuroscience, Oregon Health and Science University Portland, OR, USA ; Division of Neuroscience, Oregon National Primate Research Center Beaverton, OR, USA
| |
Collapse
|
36
|
Relaxin-3 receptor (RXFP3) signalling mediates stress-related alcohol preference in mice. PLoS One 2015; 10:e0122504. [PMID: 25849482 PMCID: PMC4388568 DOI: 10.1371/journal.pone.0122504] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022] Open
Abstract
Stressful life events are causally linked with alcohol use disorders (AUDs), providing support for a hypothesis that alcohol consumption is aimed at stress reduction. We have previously shown that expression of relaxin-3 mRNA in rat brain correlates with alcohol intake and that central antagonism of relaxin-3 receptors (RXFP3) prevents stress-induced reinstatement of alcohol-seeking. Therefore the objectives of these studies were to investigate the impact of Rxfp3 gene deletion in C57BL/6J mice on baseline and stress-related alcohol consumption. Male wild-type (WT) and Rxfp3 knockout (KO) (C57/B6JRXFP3TM1/DGen) littermate mice were tested for baseline saccharin and alcohol consumption and preference over water in a continuous access two-bottle free-choice paradigm. Another cohort of mice was subjected to repeated restraint followed by swim stress to examine stress-related alcohol preference. Hepatic alcohol and aldehyde dehydrogenase activity was assessed in mice following chronic alcohol intake and in naive controls. WT and Rxfp3 KO mice had similar baseline saccharin and alcohol preference, and hepatic alcohol processing. However, Rxfp3 KO mice displayed a stress-induced reduction in alcohol preference that was not observed in WT littermates. Notably, this phenotype, once established, persisted for at least six weeks after cessation of stress exposure. These findings suggest that in mice, relaxin-3/RXFP3 signalling is involved in maintaining high alcohol preference during and after stress, but does not appear to strongly regulate the primary reinforcing effects of alcohol.
Collapse
|
37
|
Social stress and escalated drug self-administration in mice I. Alcohol and corticosterone. Psychopharmacology (Berl) 2015; 232:991-1001. [PMID: 25242256 PMCID: PMC4339510 DOI: 10.1007/s00213-014-3733-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022]
Abstract
RATIONALE Stress experiences have been shown to be a risk factor for alcohol abuse in humans; however, a reliable mouse model using episodic social stress has yet to be developed. OBJECTIVES The current studies investigated the effects of mild and moderate social defeat protocols on plasma corticosterone, voluntary alcohol drinking, and motivation to drink alcohol. METHODS Outbred Carworth Farms Webster (CFW) mice were socially defeated for 10 days during which the intruder mouse underwent mild (15 bites: mean = 1.5 min) or moderate (30 bites: mean = 3.8 min) stress. Plasma corticosterone was measured on days 1 and 10 of the defeat. Ethanol drinking during continuous access to alcohol was measured 10 days following the defeat or 10 days prior to, during, and 20 days after the defeat. Motivation to drink was determined using a progressive ratio (PR) operant conditioning schedule during intermittent access to alcohol. RESULTS Plasma corticosterone was elevated in both stress groups on days 1 and 10. Ethanol consumption and preference following moderate stress were higher (13.3 g/kg/day intake) than both the mild stress group (8.0 g/kg/day) and controls (7.4 g/kg/day). Mice with previously acquired ethanol drinking showed decreased alcohol consumption during the moderate stress followed by an increase 20 days post-defeat. Moderately stressed mice also showed escalated ethanol intake and self-administration during a schedule of intermittent access to alcohol. CONCLUSION Social defeat experiences of moderate intensity and duration led to increased ethanol drinking and preference in CFW mice. Ongoing work investigates the interaction between glucocorticoids and dopaminergic systems as neural mechanisms for stress-escalated alcohol consumption.
Collapse
|
38
|
Acetaldehyde self-administration by a two-bottle choice paradigm: consequences on emotional reactivity, spatial learning, and memory. Alcohol 2015; 49:139-48. [PMID: 25636827 DOI: 10.1016/j.alcohol.2015.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acetaldehyde, the first alcohol metabolite, is responsible for many pharmacological effects that are not clearly distinguishable from those exerted by its parent compound. It alters motor performance, induces reinforced learning and motivated behavior, and produces different reactions according to the route of administration and the relative accumulation in the brain or in the periphery. The effective activity of oral acetaldehyde represents an unresolved field of inquiry that deserves further investigation. Thus, this study explores the acquisition and maintenance of acetaldehyde drinking behavior in adult male rats, employing a two-bottle choice paradigm for water and acetaldehyde solution (from 0.9% to 3.2% v/v), over 8 weeks. The behavioral consequences exerted by chronic acetaldehyde intake are assessed by a set of different tests: trials in an open-field arena and elevated-plus maze provided information on both general motor and explorative activity, and anxiety-driven behavioral responses. The Morris water maze allowed the exploration of cognitive processes such as spatial learning and memory. Determination of acetaldehyde levels in the brain was carried out at the end of the drinking paradigm. Our results indicate that rats exposed for the first time to acetaldehyde at 0.9% displayed a regular and stable daily drinking pattern that reached higher values and a "peaks and drops" shaped-trend when acetaldehyde concentration was increased to 3.2%. Accordingly, an increase in acetaldehyde levels in the brain was determined compared to non-acetaldehyde drinking rats. Acetaldehyde intake during the free-choice paradigm exerted an anxiogenic response in the open-field arena and elevated-plus maze, which in turn correlates with an enhancement in cognitive flexibility and spatial orientation skills, when an adaptive response to a stressful environmental challenge was required. These findings further support the idea that acetaldehyde is indeed a centrally active and behaviorally relevant metabolite of alcohol.
Collapse
|
39
|
Phillips TJ, Reed C, Pastor R. Preclinical evidence implicating corticotropin-releasing factor signaling in ethanol consumption and neuroadaptation. GENES, BRAIN, AND BEHAVIOR 2015; 14:98-135. [PMID: 25565358 PMCID: PMC4851463 DOI: 10.1111/gbb.12189] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/22/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
The results of many studies support the influence of the corticotropin-releasing factor (CRF) system on ethanol (EtOH) consumption and EtOH-induced neuroadaptations that are critical in the addiction process. This review summarizes the preclinical data in this area after first providing an overview of the components of the CRF system. This complex system involves hypothalamic and extra-hypothalamic mechanisms that play a role in the central and peripheral consequences of stressors, including EtOH and other drugs of abuse. In addition, several endogenous ligands and targets make up this system and show differences in their involvement in EtOH drinking and in the effects of chronic or repeated EtOH treatment. In general, genetic and pharmacological approaches paint a consistent picture of the importance of CRF signaling via type 1 CRF receptors (CRF(1)) in EtOH-induced neuroadaptations that result in higher levels of intake, encourage alcohol seeking during abstinence and alter EtOH sensitivity. Furthermore, genetic findings in rodents, non-human primates and humans have provided some evidence of associations of genetic polymorphisms in CRF-related genes with EtOH drinking, although additional data are needed. These results suggest that CRF(1) antagonists have potential as pharmacotherapeutics for alcohol use disorders. However, given the broad and important role of these receptors in adaptation to environmental and other challenges, full antagonist effects may be too profound and consideration should be given to treatments with modulatory effects.
Collapse
Affiliation(s)
- T. J. Phillips
- VA Portland Health Care System, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - C. Reed
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - R. Pastor
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
- Area de Psicobiología, Universitat Jaume I, 12071 Castellón, Spain
- Department of Psychology, Reed College, Portland, OR, USA
| |
Collapse
|
40
|
CRF1 receptor-deficiency induces anxiety-like vulnerability to cocaine. Psychopharmacology (Berl) 2014; 231:3965-72. [PMID: 24687410 DOI: 10.1007/s00213-014-3534-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/03/2014] [Indexed: 01/23/2023]
Abstract
RATIONALE The intake of psychostimulant drugs may induce cognitive dysfunction and negative affective-like states, and is associated with increased activity of stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates neuroendocrine, behavioural and autonomic responses to stressors, and might be implicated in substance-related disorders. CRF signalling is mediated by two receptor types, named CRF1 and CRF2. OBJECTIVES The present study aims to elucidate the role for the CRF1 receptor in cognitive dysfunction and anxiety-like states induced by cocaine. RESULTS The genetic inactivation of the CRF1 receptor (CRF1+/- and CRF1-/-) does not influence recognition memory in drug-naïve mice, as assessed by the novel object recognition (NOR) test. Moreover, the chronic administration of escalating doses of cocaine (5-20 mg/kg, i.p.) induces NOR deficits, which are unaffected by CRF1 receptor-deficiency. However, the same drug regimen reveals an anxiety-like vulnerability to cocaine in CRF1-/- but not in wild-type or CRF1+/- mice, as assessed by the elevated plus maze test. CONCLUSIONS The present findings indicate dissociation of cognitive dysfunction and anxiety-like states induced by cocaine. Moreover, they unravel a novel mechanism of vulnerability to psychostimulant drugs.
Collapse
|
41
|
Leonelli S, Ankeny RA, Nelson NC, Ramsden E. Making organisms model human behavior: situated models in North-American alcohol research, since 1950. SCIENCE IN CONTEXT 2014; 27:485-509. [PMID: 25233743 PMCID: PMC4274764 DOI: 10.1017/s0269889714000155] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We examine the criteria used to validate the use of nonhuman organisms in North-American alcohol addiction research from the 1950s to the present day. We argue that this field, where the similarities between behaviors in humans and non-humans are particularly difficult to assess, has addressed questions of model validity by transforming the situatedness of non-human organisms into an experimental tool. We demonstrate that model validity does not hinge on the standardization of one type of organism in isolation, as often the case with genetic model organisms. Rather, organisms are viewed as necessarily situated: they cannot be understood as a model for human behavior in isolation from their environmental conditions. Hence the environment itself is standardized as part of the modeling process; and model validity is assessed with reference to the environmental conditions under which organisms are studied.
Collapse
Affiliation(s)
| | - Rachel A. Ankeny
- School of History & Politics, University of Adelaide, Napier 423, Adelaide 5005 SA, Australia,
| | - Nicole C. Nelson
- Department of Social Studies of Medicine, McGill University, 3647 Peel Room 207, Montreal QC, H3A 1X1, Canada,
| | - Edmund Ramsden
- Centre for the History of Science, Technology and Medicine, Faculty of Life Sciences, University of Manchester, Simon Building, Manchester, M13 9PL, UK
| |
Collapse
|
42
|
Jeanblanc J, Coune F, Botia B, Naassila M. Brain-derived neurotrophic factor mediates the suppression of alcohol self-administration by memantine. Addict Biol 2014; 19:758-69. [PMID: 23414063 DOI: 10.1111/adb.12039] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) within the striatum is part of a homeostatic pathway regulating alcohol consumption. Memantine, a non-competitive antagonist of N-methyl-D-aspartate receptors, induces expression of BDNF in several brain regions including the striatum. We hypothesized that memantine could decrease ethanol (EtOH) consumption via activation of the BNDF signalling pathway. Effects of memantine were evaluated in Long-Evans rats self-administering moderate or high amounts of EtOH 6, 30 and 54 hours after an acute injection (12.5 and 25 mg/kg). Motivation to consume alcohol was investigated through a progressive ratio paradigm. The possible role for BDNF in the memantine effect was tested by blockade of the TrkB receptor using the pharmacological agent K252a and by the BDNF scavenger TrkB-Fc. Candidate genes expression was also assessed by polymerase chain reaction array 4 and 28 hours after memantine injection. We found that memantine decreased EtOH self-administration and motivation to consume EtOH 6 and 30 hours post-injection. In addition, we found that inhibition or blockade of the BDNF signalling pathway prevented the early, but not the delayed decrease in EtOH consumption induced by memantine. Finally, Bdnf expression was differentially regulated between the early and delayed timepoints. These results demonstrate that an acute injection of memantine specifically reduces EtOH self-administration and motivation to consume EtOH for at least 30 hours. Moreover, we showed that BDNF was responsible for the early effect, but that the delayed effect was BDNF-independent.
Collapse
Affiliation(s)
- Jérôme Jeanblanc
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances - INSERM ERI 24; UFR de Pharmacie; Université de Picardie Jules Verne, SFR CAP Santé; France
| | - Fabien Coune
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances - INSERM ERI 24; UFR de Pharmacie; Université de Picardie Jules Verne, SFR CAP Santé; France
| | - Béatrice Botia
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances - INSERM ERI 24; UFR de Pharmacie; Université de Picardie Jules Verne, SFR CAP Santé; France
| | - Mickaël Naassila
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances - INSERM ERI 24; UFR de Pharmacie; Université de Picardie Jules Verne, SFR CAP Santé; France
| |
Collapse
|
43
|
Spanagel R, Noori HR, Heilig M. Stress and alcohol interactions: animal studies and clinical significance. Trends Neurosci 2014; 37:219-27. [PMID: 24636458 DOI: 10.1016/j.tins.2014.02.006] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 02/04/2014] [Accepted: 02/07/2014] [Indexed: 01/05/2023]
Abstract
Alcohol is frequently consumed for stress relief, but the individual determinants and the temporal course of stress-induced alcohol use are not well understood. Preclinical studies may help shed light on these factors. We synthesize here the findings from numerous rodent studies of stress and alcohol interactions. Stress-induced alcohol consumption is age-dependent, has a high genetic load, and results from an interaction of the stress and reward systems. Specifically, glucocorticoids, acting within the nucleus accumbens (NAc), are important mediators of this stress-induced alcohol intake. In addition, increased activation of the corticotropin-releasing hormone (CRH) system within the extended amygdala appears to mediate stress-induced relapse. Finally, these preclinical studies have helped to identify several attractive targets for novel treatments of alcohol abuse and addiction.
Collapse
Affiliation(s)
- Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health (CIMH), University of Heidelberg, Medical Faculty Mannheim, Germany
| | - Hamid R Noori
- Institute of Psychopharmacology, Central Institute of Mental Health (CIMH), University of Heidelberg, Medical Faculty Mannheim, Germany
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, USA
| |
Collapse
|
44
|
Legastelois R, Botia B, Coune F, Jeanblanc J, Naassila M. Deciphering the relationship between vulnerability to ethanol-induced behavioral sensitization and ethanol consumption in outbred mice. Addict Biol 2014; 19:210-24. [PMID: 24164956 DOI: 10.1111/adb.12104] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Ethanol (EtOH)-induced behavioral sensitization (EIBS) is proposed to play a role in early and recurring steps of alcohol dependence, but its impact on alcohol abuse is not clear. EIBS development is dependent upon animal species, strain and also individual factors. We proposed here to decipher the co-expression of EIBS and EtOH intake in individual animals among outbred Swiss mice, which exhibit heterogeneity that parallels what may occur in humans. To do so, mice were exposed to a two-bottle choice with free access to water or 10% EtOH for 6 days just before and immediately after chronic intraperitoneal 2.5 g/kg ethanol injections once a day for 10 consecutive days. Based on their sensitization scores, mice were split into resistant and sensitized animals. First, we showed that individual susceptibility to EIBS is inversely correlated with voluntary EtOH consumption. Exposure to repeated EtOH during EIBS development increased subsequent EtOH intake among the entire population. Very interestingly, subsequent analyses suggested that the less the mice are sensitized the more they increase their EtOH intake; however, resistant mice were sensitive to EtOH adulteration with quinine, whereas sensitized ones maintained their EtOH intake levels, therefore exhibiting a compulsive-like drinking pattern. In addition, we showed that resistant mice do not exhibit a weaker sensitivity to the aversive properties of EtOH that may contribute to their higher level of EtOH intake compared to sensitized mice. This study confirms and extends previous data showing a deep relationship between propensity for EtOH consumption and susceptibility to EIBS in Swiss mice.
Collapse
Affiliation(s)
- Rémi Legastelois
- Université de Picardie Jules Verne; UFR Pharmacie; INSERM Unit ERi 24 GRAP France
| | - Béatrice Botia
- Université de Picardie Jules Verne; UFR Pharmacie; INSERM Unit ERi 24 GRAP France
| | - Fabien Coune
- Université de Picardie Jules Verne; UFR Pharmacie; INSERM Unit ERi 24 GRAP France
| | - Jérôme Jeanblanc
- Université de Picardie Jules Verne; UFR Pharmacie; INSERM Unit ERi 24 GRAP France
| | - Mickaël Naassila
- Université de Picardie Jules Verne; UFR Pharmacie; INSERM Unit ERi 24 GRAP France
| |
Collapse
|
45
|
Abstract
Chronic consumption of a large amount of alcohol disrupts the communication between nervous, endocrine, and immune system and causes hormonal disturbances that lead to profound and serious consequences at physiologic and behavioral levels. These alcohol-induced hormonal dysregulations affect the entire body and can result in various disorders such as stress abnormalities, reproductive deficits, body growth defect, thyroid problems, immune dysfunction, cancers, bone disease, and psychological and behavioral disorders. This review summarizes the findings from human and animal studies that provide consistent evidence on the various effects of alcohol abuse on the endocrine system.
Collapse
Affiliation(s)
- Nadia Rachdaoui
- Nadia Rachdaoui, Ph.D., Rutgers Endocrine Research Program. Department of Animal Sciences Rutgers University, 67 Poultry Farm Lane, New Brunswick, NJ 08901,
| | - Dipak K. Sarkar
- Dipak K. Sarkar, Ph.D., D. Phil., Rutgers Endocrine Research Program. Department of Animal Sciences, Rutgers University, 67 Poultry Farm Lane, New Brunswick, NJ 08901,
| |
Collapse
|
46
|
Kratzer S, Mattusch C, Metzger MW, Dedic N, Noll-Hussong M, Kafitz KW, Eder M, Deussing JM, Holsboer F, Kochs E, Rammes G. Activation of CRH receptor type 1 expressed on glutamatergic neurons increases excitability of CA1 pyramidal neurons by the modulation of voltage-gated ion channels. Front Cell Neurosci 2013; 7:91. [PMID: 23882180 PMCID: PMC3715697 DOI: 10.3389/fncel.2013.00091] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 05/25/2013] [Indexed: 01/16/2023] Open
Abstract
Corticotropin-releasing hormone (CRH) plays an important role in a substantial number of patients with stress-related mental disorders, such as anxiety disorders and depression. CRH has been shown to increase neuronal excitability in the hippocampus, but the underlying mechanisms are poorly understood. The effects of CRH on neuronal excitability were investigated in acute hippocampal brain slices. Population spikes (PS) and field excitatory postsynaptic potentials (fEPSP) were evoked by stimulating Schaffer-collaterals and recorded simultaneously from the somatic and dendritic region of CA1 pyramidal neurons. CRH was found to increase PS amplitudes (mean ± Standard error of the mean; 231.8 ± 31.2% of control; n = 10) while neither affecting fEPSPs (104.3 ± 4.2%; n = 10) nor long-term potentiation (LTP). However, when Schaffer-collaterals were excited via action potentials (APs) generated by stimulation of CA3 pyramidal neurons, CRH increased fEPSP amplitudes (119.8 ± 3.6%; n = 8) and the magnitude of LTP in the CA1 region. Experiments in slices from transgenic mice revealed that the effect on PS amplitude is mediated exclusively by CRH receptor 1 (CRHR1) expressed on glutamatergic neurons. The effects of CRH on PS were dependent on phosphatase-2B, L- and T-type calcium channels and voltage-gated potassium channels but independent on intracellular Ca2+-elevation. In patch-clamp experiments, CRH increased the frequency and decay times of APs and decreased currents through A-type and delayed-rectifier potassium channels. These results suggest that CRH does not affect synaptic transmission per se, but modulates voltage-gated ion currents important for the generation of APs and hence elevates by this route overall neuronal activity.
Collapse
Affiliation(s)
- Stephan Kratzer
- Department of Anesthesiology, Klinikum Rechts der Isar der Technischen Universität München Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Giardino WJ, Ryabinin AE. CRF1 receptor signaling regulates food and fluid intake in the drinking-in-the-dark model of binge alcohol consumption. Alcohol Clin Exp Res 2013; 37:1161-70. [PMID: 23398267 DOI: 10.1111/acer.12076] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/30/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND Several recent studies implementing the standard "drinking-in-the-dark" (DID) model of short-term binge-like ethanol (EtOH) intake in C57BL/6J mice highlighted a role for the stress-related neuropeptide corticotropin-releasing factor (CRF) and its primary binding partner, the CRF type-1 (CRF1) receptor. METHODS We evaluated the selectivity of CRF1 involvement in binge-like EtOH intake by interrupting CRF1 function via pharmacological and genetic methods in a slightly modified 2-bottle choice DID model that allowed calculation of an EtOH preference ratio. In addition to determining EtOH intake and preference, we also measured consumption of food and H2 O during the DID period, both in the presence and absence of EtOH and sweet tastant solutions. RESULTS Treatment with either of the CRF1-selective antagonists CP-376,395 (CP; 10 to 20 mg/kg, i.p.) or NBI-27914 (10 to 30 mg/kg, i.p.) decreased intake of 15% EtOH in male C57BL/6J mice, but did so in the absence of a concomitant decrease in EtOH preference. These findings were replicated genetically in a CRF1 knockout (KO) mouse model (also on a C57BL/6J background). In contrast to effects on EtOH intake, pharmacological blockade of CRF1 with CP increased intake of 10% sucrose, consistent with previous findings in CRF1 KO mice. Finally, pharmacological and genetic disruption of CRF1 activity significantly reduced feeding and/or total caloric intake in all experiments, confirming the existence of nonspecific effects. CONCLUSIONS Our findings indicate that blockade of CRF1 receptors does not exert specific effects on EtOH intake in the DID paradigm, and that slight modifications to this procedure, as well as additional consummatory control experiments, may be useful when evaluating the selectivity of pharmacological and genetic manipulations on binge-like EtOH intake.
Collapse
Affiliation(s)
- William J Giardino
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|
48
|
Bardo MT, Neisewander JL, Kelly TH. Individual differences and social influences on the neurobehavioral pharmacology of abused drugs. Pharmacol Rev 2013; 65:255-90. [PMID: 23343975 PMCID: PMC3565917 DOI: 10.1124/pr.111.005124] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The interaction of drugs with biologic targets is a critical area of research, particularly for the development of medications to treat substance use disorders. In addition to understanding these drug-target interactions, however, there is a need to understand more fully the psychosocial influences that moderate these interactions. The first section of this review introduces some examples from human behavioral pharmacology that illustrate the clinical importance of this research. The second section covers preclinical evidence to characterize some of the key individual differences that alter drug sensitivity and abuse vulnerability, related primarily to differences in response to novelty and impulsivity. Evidence is presented to indicate that critical neuropharmacological mechanisms associated with these individual differences involve integrated neurocircuits underlying stress, reward, and behavioral inhibitory processes. The third section covers social influences on drug abuse vulnerability, including effects experienced during infancy, adolescence, and young adulthood, such as maternal separation, housing conditions, and social interactions (defeat, play, and social rank). Some of the same neurocircuits involved in individual differences also are altered by social influences, although the precise neurochemical and cellular mechanisms involved remain to be elucidated fully. Finally, some speculation is offered about the implications of this research for the prevention and treatment of substance abuse.
Collapse
Affiliation(s)
- M T Bardo
- Department of Psychology, University of Kentucky, BBSRB Room 447, 741 S. Limestone, Lexington, KY 40536-0509, USA.
| | | | | |
Collapse
|
49
|
Maul B, Becker M, Gembardt F, Becker A, Schultheiss HP, Siems WE, Walther T. Genetic deficiency in neprilysin or its pharmacological inhibition initiate excessive stress-induced alcohol consumption in mice. PLoS One 2012. [PMID: 23185571 PMCID: PMC3503995 DOI: 10.1371/journal.pone.0050187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Both acquired and inherited genetic factors contribute to excessive alcohol consumption and the corresponding development of addiction. Here we show that the genetic deficiency in neprilysin [NEP] did not change the kinetics of alcohol degradation but led to an increase in alcohol intake in mice in a 2-bottle-free-choice paradigm after one single stress stimulus (intruder). A repetition of such stress led to an irreversible elevated alcohol consumption. This phenomenon could be also observed in wild-type mice receiving an orally active NEP inhibitor. We therefore elucidated the stress behavior in NEP-deficient mice. In an Elevated Plus Maze, NEP knockouts crossed more often the area between the arms, implicating a significant stronger stress response. Furthermore, such animals showed a decreased locomotor activity under intense light in a locomotor activity test, identifying such mice to be more responsive in aversive situations than their wild-type controls. Since the reduction in NEP activity itself does not lead to significant signs of an altered alcohol preference in mice but requires an environmental stimulus, our findings build a bridge between stress components and genetic factors in the development of alcoholism. Therefore, targeting NEP activity might be a very attractive approach for the treatment of alcohol abuse in a society with increasing social and financial stress.
Collapse
Affiliation(s)
- Björn Maul
- Department for Biochemical Neurobiology, Leibnizinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Matthias Becker
- Department for Biochemical Neurobiology, Leibnizinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Florian Gembardt
- Department of Cardiac Pathobiology, Excellence Cluster Cardio-Pulmonary System, Giessen, Germany
- Department for Nephrology – MK3, University Hospital Dresden, Dresden, Germany
| | - Axel Becker
- Institute of Pharmacology and Toxicology, Otto-von-Guericke-University of Magdeburg, Magdeburg, Germany
| | | | - Wolf-Eberhard Siems
- Department for Biochemical Neurobiology, Leibnizinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Thomas Walther
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim – University Heidelberg, Mannheim, Germany
- Centre for Fetal Medicine, Department of Pediatric Surgery & Department of Obstetrics, Division of Women and Child Health, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
50
|
Abstract
Ethanol (EtOH) has effects on numerous cellular molecular targets, and alterations in synaptic function are prominent among these effects. Acute exposure to EtOH activates or inhibits the function of proteins involved in synaptic transmission, while chronic exposure often produces opposing and/or compensatory/homeostatic effects on the expression, localization, and function of these proteins. Interactions between different neurotransmitters (e.g., neuropeptide effects on release of small molecule transmitters) can also influence both acute and chronic EtOH actions. Studies in intact animals indicate that the proteins affected by EtOH also play roles in the neural actions of the drug, including acute intoxication, tolerance, dependence, and the seeking and drinking of EtOH. This chapter reviews the literature describing these acute and chronic synaptic effects of EtOH and their relevance for synaptic transmission, plasticity, and behavior.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, NIAAA, 5625 Fishers Lane, Room TS-13A, Rockville, MD 20852, USA.
| | | |
Collapse
|