1
|
Neel AI, Wang Y, Sun H, Liontis KE, McCormack MC, Mayer JC, Cervera Juanes RP, Davenport AT, Grant KA, Daunais JD, Chen R. Differential regulation of G protein-coupled receptor-associated proteins in the caudate and the putamen of cynomolgus macaques following chronic ethanol drinking. J Neurochem 2024; 168:2722-2735. [PMID: 38783749 PMCID: PMC11449652 DOI: 10.1111/jnc.16134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/16/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
The dorsal striatum is composed of the caudate nucleus and the putamen in human and non-human primates. These two regions receive different cortical projections and are functionally distinct. The caudate is involved in the control of goal-directed behaviors, while the putamen is implicated in habit learning and formation. Previous reports indicate that ethanol differentially influences neurotransmission in these two regions. Because neurotransmitters primarily signal through G protein-coupled receptors (GPCRs) to modulate neuronal activity, the present study aimed to determine whether ethanol had a region-dependent impact on the expression of proteins that are involved in the trafficking and function of GPCRs, including G protein subunits and their effectors, protein kinases, and elements of the cytoskeleton. Western blotting was performed to examine protein levels in the caudate and the putamen of male cynomolgus macaques that self-administered ethanol for 1 year under free access conditions, along with control animals that self-administered an isocaloric sweetened solution under identical operant conditions. Among the 18 proteins studied, we found that the levels of one protein (PKCβ) were increased, and 13 proteins (Gαi1/3, Gαi2, Gαo, Gβ1γ, PKCα, PKCε, CaMKII, GSK3β, β-actin, cofilin, α-tubulin, and tubulin polymerization promoting protein) were reduced in the caudate of alcohol-drinking macaques. However, ethanol did not alter the expression of any proteins examined in the putamen. These observations underscore the unique vulnerability of the caudate nucleus to changes in protein expression induced by chronic ethanol exposure. Whether these alterations are associated with ethanol-induced dysregulation of GPCR function and neurotransmission warrants future investigation.
Collapse
Affiliation(s)
- Anna I. Neel
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Yutong Wang
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Haiguo Sun
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Katherine E. Liontis
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Mary C. McCormack
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Jonathan C. Mayer
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Rita P. Cervera Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - April T. Davenport
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Kathleen A. Grant
- Division of Neuroscience Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR 97239
| | - James D. Daunais
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Rong Chen
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| |
Collapse
|
2
|
Favoretto CA, Bertagna NB, Miguel TT, Quadros IMH. The CRF/Urocortin systems as therapeutic targets for alcohol use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:97-152. [PMID: 39523064 DOI: 10.1016/bs.irn.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Development and maintenance of alcohol use disorders have been proposed to recruit critical mechanisms involving Corticotropin Releasing Factor and Urocortins (CRF/Ucns). The CRF/Ucns system is comprised of a family of peptides (CRF, Ucn 1, Ucn 2, Ucn 3) which act upon two receptor subtypes, CRFR1 and CRFR2, each with different affinity profiles to the endogenous peptides and differential brain distribution. Activity of CRF/Ucn system is further modulated by CRF binding protein (CRF-BP), which regulates availability of CRF and Ucns to exert their actions. Extensive evidence in preclinical models support the involvement of CRF/Ucn targets in escalated alcohol drinking, as well as point to changes in CRF/Ucn brain function as a result of chronic alcohol exposure and/or withdrawal. It highlights the role of CRF and CRFR1-mediated signaling in conditions of excessive alcohol taking and seeking, including during various stages of withdrawal and relapse to alcohol. Besides its role in the hypothalamic-pituitary-adrenal (HPA) axis, the importance of extra-hypothalamic CRF pathways, especially in the extended amygdala, in the neurobiology of alcohol abuse and dependence is emphasized. Emerging roles for other targets of the CRF/Ucn system, such as CRF2 receptors, CRF-BP and Ucns in escalated alcohol drinking is also discussed. Finally, the limited translational value of CRF/Ucn interventions in stress-related and alcohol use disorders is discussed. So far, CRFR1 antagonists have shown little or no efficacy in human clinical trials, although a range of unexplored conditions and possibilities remain to be explored.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Natalia Bonetti Bertagna
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil
| | | | - Isabel M H Quadros
- Psychobiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil.
| |
Collapse
|
3
|
Gimenez-Gomez P, Le T, Martin GE. Modulation of neuronal excitability by binge alcohol drinking. Front Mol Neurosci 2023; 16:1098211. [PMID: 36866357 PMCID: PMC9971943 DOI: 10.3389/fnmol.2023.1098211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
Drug use poses a serious threat to health systems throughout the world. The number of consumers rises every year being alcohol the drug of abuse most consumed causing 3 million deaths (5.3% of all deaths) worldwide and 132.6 million disability-adjusted life years. In this review, we present an up-to-date summary about what is known regarding the global impact of binge alcohol drinking on brains and how it affects the development of cognitive functions, as well as the various preclinical models used to probe its effects on the neurobiology of the brain. This will be followed by a detailed report on the state of our current knowledge of the molecular and cellular mechanisms underlying the effects of binge drinking on neuronal excitability and synaptic plasticity, with an emphasis on brain regions of the meso-cortico limbic neurocircuitry.
Collapse
Affiliation(s)
- Pablo Gimenez-Gomez
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| | - Timmy Le
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, United States
| | - Gilles E. Martin
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| |
Collapse
|
4
|
Kreifeldt M, Herman MA, Sidhu H, Okhuarobo A, Macedo GC, Shahryari R, Gandhi PJ, Roberto M, Contet C. Central amygdala corticotropin-releasing factor neurons promote hyponeophagia but do not control alcohol drinking in mice. Mol Psychiatry 2022; 27:2502-2513. [PMID: 35264727 PMCID: PMC9149056 DOI: 10.1038/s41380-022-01496-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 12/20/2022]
Abstract
Corticotropin-releasing factor (CRF) signaling in the central nucleus of the amygdala (CeA) plays a critical role in rodent models of excessive alcohol drinking. However, the source of CRF acting in the CeA during alcohol withdrawal remains to be identified. In the present study, we hypothesized that CeA CRF interneurons may represent a behaviorally relevant source of CRF to the CeA increasing motivation for alcohol via negative reinforcement. We first observed that Crh mRNA expression in the anterior part of the mouse CeA correlates positively with alcohol intake in C57BL/6J males with a history of chronic binge drinking followed by abstinence and increases upon exposure to chronic intermittent ethanol (CIE) vapor inhalation. We then found that chemogenetic activation of CeA CRF neurons in Crh-IRES-Cre mouse brain slices increases gamma-aminobutyric acid (GABA) release in the medial CeA, in part via CRF1 receptor activation. While chemogenetic stimulation exacerbated novelty-induced feeding suppression (NSF) in alcohol-naïve mice, thereby mimicking the effect of withdrawal from CIE, it had no effect on voluntary alcohol consumption, following either acute or chronic manipulation. Furthermore, chemogenetic inhibition of CeA CRF neurons did not affect alcohol consumption or NSF in chronic alcohol drinkers exposed to air or CIE. Altogether, these findings indicate that CeA CRF neurons produce local release of GABA and CRF and promote hyponeophagia in naïve mice, but do not drive alcohol intake escalation or negative affect in CIE-withdrawn mice. The latter result contrasts with previous findings in rats and demonstrates species specificity of CRF circuit engagement in alcohol dependence.
Collapse
Affiliation(s)
- Max Kreifeldt
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Melissa A Herman
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Harpreet Sidhu
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Agbonlahor Okhuarobo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
- University of Benin, Faculty of Pharmacy, Department of Pharmacology & Toxicology, Benin City, Nigeria
| | - Giovana C Macedo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Roxana Shahryari
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Pauravi J Gandhi
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA.
| |
Collapse
|
5
|
Agoglia AE, Zhu M, Quadir SG, Bluitt MN, Douglass E, Hanback T, Tella J, Ying R, Hodge CW, Herman MA. Sex-specific plasticity in CRF regulation of inhibitory control in central amygdala CRF1 neurons after chronic voluntary alcohol drinking. Addict Biol 2022; 27:e13067. [PMID: 34075665 PMCID: PMC8636550 DOI: 10.1111/adb.13067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/01/2021] [Accepted: 05/14/2021] [Indexed: 01/03/2023]
Abstract
Despite strong preclinical evidence for the ability of corticotropin releasing factor 1 (CRF1) antagonists to regulate alcohol consumption, clinical trials have not yet demonstrated therapeutic effects of these compounds in alcohol use disorder (AUD) patients. Several confounding factors may limit the translation of preclinical CRF1 research to patients, including reliance on experimenter-administered alcohol instead of voluntary consumption, a preponderance of evidence collected in male subjects only and an inability to assess the effects of alcohol on specific brain circuits. A population of particular interest is the CRF1-containing neurons of the central amygdala (CeA). CRF1 CeA neurons are sensitive to ethanol, but the effects of alcohol drinking on CRF signalling within this population are unknown. In the present study, we assessed the effects of voluntary alcohol drinking on inhibitory control of CRF1+ CeA neurons from male and female CRF1:GFP mice using ex vivo electrophysiology and determined the contributions of CRF1 signalling to inhibitory control and voluntary alcohol drinking. Chronic alcohol drinking produced neuroadaptations in CRF1+ neurons that increased the sensitivity of GABAA receptor-mediated sIPSCs to the acute effects of alcohol, CRF and the CRF1 antagonist R121919, but these adaptations were more pronounced in male versus female mice. The CRF1 antagonist CP-154,526 reduced voluntary alcohol drinking in both sexes and abolished sex differences in alcohol drinking. The lack of alcohol-induced adaptation in the female CRF1 system may be related to the elevated alcohol intake exhibited by female mice and could contribute to the ineffectiveness of CRF1 antagonists in female AUD patients.
Collapse
Affiliation(s)
- AE Agoglia
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - M Zhu
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - SG Quadir
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - MN Bluitt
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - E Douglass
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - T Hanback
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - J Tella
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - R Ying
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - CW Hodge
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - MA Herman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
6
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
7
|
Li BX, Dong GH, Li HL, Zhang JS, Bing YH, Chu CP, Cui SB, Qiu DL. Chronic Ethanol Exposure Enhances Facial Stimulation-Evoked Mossy Fiber-Granule Cell Synaptic Transmission via GluN2A Receptors in the Mouse Cerebellar Cortex. Front Syst Neurosci 2021; 15:657884. [PMID: 34408633 PMCID: PMC8365521 DOI: 10.3389/fnsys.2021.657884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022] Open
Abstract
Sensory information is transferred to the cerebellar cortex via the mossy fiber–granule cell (MF–GC) pathway, which participates in motor coordination and motor learning. We previously reported that chronic ethanol exposure from adolescence facilitated the sensory-evoked molecular layer interneuron–Purkinje cell synaptic transmission in adult mice in vivo. Herein, we investigated the effect of chronic ethanol exposure from adolescence on facial stimulation-evoked MF–GC synaptic transmission in the adult mouse cerebellar cortex using electrophysiological recording techniques and pharmacological methods. Chronic ethanol exposure from adolescence induced an enhancement of facial stimulation-evoked MF–GC synaptic transmission in the cerebellar cortex of adult mice. The application of an N-methyl-D-aspartate receptor (NMDAR) antagonist, D-APV (250 μM), induced stronger depression of facial stimulation-evoked MF–GC synaptic transmission in chronic ethanol-exposed mice compared with that in control mice. Chronic ethanol exposure-induced facilitation of facial stimulation evoked by MF–GC synaptic transmission was abolished by a selective GluN2A antagonist, PEAQX (10 μM), but was unaffected by the application of a selective GluN2B antagonist, TCN-237 (10 μM), or a type 1 metabotropic glutamate receptor blocker, JNJ16259685 (10 μM). These results indicate that chronic ethanol exposure from adolescence enhances facial stimulation-evoked MF–GC synaptic transmission via GluN2A, which suggests that chronic ethanol exposure from adolescence impairs the high-fidelity transmission capability of sensory information in the cerebellar cortex by enhancing the NMDAR-mediated components of MF–GC synaptic transmission in adult mice in vivo.
Collapse
Affiliation(s)
- Bing-Xue Li
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Guang-Hui Dong
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Hao-Long Li
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Jia-Song Zhang
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Yan-Hua Bing
- Brain Science Research Center, Yanbian University, Yanji, China
| | - Chun-Ping Chu
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Song-Biao Cui
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - De-Lai Qiu
- Brain Science Research Center, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
8
|
Xu H, Li H, Liu D, Wen W, Xu M, Frank JA, Chen J, Zhu H, Grahame NJ, Luo J. Chronic Voluntary Alcohol Drinking Causes Anxiety-like Behavior, Thiamine Deficiency, and Brain Damage of Female Crossed High Alcohol Preferring Mice. Front Pharmacol 2021; 12:614396. [PMID: 33767622 PMCID: PMC7985542 DOI: 10.3389/fphar.2021.614396] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
The central nervous system is vulnerable to chronic alcohol abuse, and alcohol dependence is a chronically relapsing disorder which causes a variety of physical and mental disorders. Appropriate animal models are important for investigating the underlying cellular and molecular mechanisms. The crossed High Alcohol Preferring mice prefer alcohol to water when given free access. In the present study, we used female cHAP mice as a model of chronic voluntary drinking to evaluate the effects of alcohol on neurobehavioral and neuropathological changes. The female cHAP mice had free-choice access to 10% ethanol and water, while control mice had access to water alone at the age of 60-day-old. The mice were exposed to alcohol for 7 months then subjected to neurobehavioral tests including open field (OF), elevated plus maze (EPM), and Morris water maze (MWM). Results from OF and EPM tests suggested that chronic voluntary drinking caused anxiety-like behaviors. After behavior tests, mice were sacrificed, and brain tissues were processed for biochemical analyses. Alcohol altered the levels of several neurotransmitters and neurotrophic factors in the brain including gamma-Aminobutyric acid (GABA), corticotropin-releasing factor, cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor. Alcohol increased the expression of neuroinflammation markers including interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1) and C-C chemokine receptor 2 (CCR2). Alcohol also induced cleaved caspase-3 and glial fibrillary acidic protein, indicative of neurodegeneration and gliosis. In addition, alcohol inhibited the expression of thiamine transporters in the brain and reduced thiamine levels in the blood. Alcohol also caused oxidative stress and endoplasmic reticulum (ER) stress, and stimulated neurogenesis.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Hui Li
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Dexiang Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, China
| | - Wen Wen
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jacqueline A Frank
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Nicholas J Grahame
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Jia Luo
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, United States.,Iowa City VA Health Care System, Iowa City, IA, United States
| |
Collapse
|
9
|
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central nucleus of the amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptive mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid (GABA)ergic transmission in the CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the CeA, whereas chronic alcohol up-regulates NMDA receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and antistress (e.g., nociceptin/orphanin FQ, oxytocin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Dean Kirson
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
10
|
Cofresí RU, Bartholow BD, Piasecki TM. Evidence for incentive salience sensitization as a pathway to alcohol use disorder. Neurosci Biobehav Rev 2019; 107:897-926. [PMID: 31672617 PMCID: PMC6878895 DOI: 10.1016/j.neubiorev.2019.10.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
The incentive salience sensitization (ISS) theory of addiction holds that addictive behavior stems from the ability of drugs to progressively sensitize the brain circuitry that mediates attribution of incentive salience (IS) to reward-predictive cues and its behavioral manifestations. In this article, we establish the plausibility of ISS as an etiological pathway to alcohol use disorder (AUD). We provide a comprehensive and critical review of evidence for: (1) the ability of alcohol to sensitize the brain circuitry of IS attribution and expression; and (2) attribution of IS to alcohol-predictive cues and its sensitization in humans and non-human animals. We point out gaps in the literature and how these might be addressed. We also highlight how individuals with different alcohol subjective response phenotypes may differ in susceptibility to ISS as a pathway to AUD. Finally, we discuss important implications of this neuropsychological mechanism in AUD for psychological and pharmacological interventions attempting to attenuate alcohol craving and cue reactivity.
Collapse
Affiliation(s)
- Roberto U Cofresí
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States.
| | - Bruce D Bartholow
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| | - Thomas M Piasecki
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| |
Collapse
|
11
|
Centanni SW, Bedse G, Patel S, Winder DG. Driving the Downward Spiral: Alcohol-Induced Dysregulation of Extended Amygdala Circuits and Negative Affect. Alcohol Clin Exp Res 2019; 43:2000-2013. [PMID: 31403699 PMCID: PMC6779502 DOI: 10.1111/acer.14178] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) afflicts a large number of individuals, families, and communities globally. Affective disturbances, including stress, depression, and anxiety, are highly comorbid with AUD, contributing in some cases to initial alcohol use and continued use. Negative affect has a particularly strong influence on the withdrawal/abstinence stage of addiction as individuals with AUD frequently report stressful events, depression, and anxiety as key factors for relapse. Treatment options for negative affect associated with AUD are limited and often ineffective, highlighting the pressing need for preclinical studies examining the underlying neural circuitry driving AUD-associated negative affect. The extended amygdala (EA) is a set of brain areas collectively involved in generating and regulating affect, and extensive research has defined a critical role for the EA in all facets of substance use disorder. Here, we review the expansive historical literature examining the effects of ethanol exposure on the EA, with an emphasis on the complex EA neural circuitry driving negative affect in all phases of the alcohol addiction cycle. Specifically, this review focuses on the effects of alcohol exposure on the neural circuitry in 2 key components of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis. Additionally, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and neural circuitry in the EA, with the long-term goal of developing better diagnostic tools and new pharmacological targets aimed at treating negative affect in AUD. The concepts detailed here will serve as the foundation for a companion review focusing on the potential for the endogenous cannabinoid system in the EA as a novel target for treating the stress, anxiety, and negative emotional state driving AUD.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
12
|
Li LB, Kim YW, Wang YH, Bai L, Zhu XD, Zhao ZL, Lee CW, Jiao Y, Wu T, Cai ZZ, Kim SC, An WG, Yang CH, Cui GC, Zhao RJ. Methanol extract of semen Ziziphi Spinosae attenuates ethanol withdrawal anxiety by improving neuropeptide signaling in the central amygdala. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:147. [PMID: 31234859 PMCID: PMC6591875 DOI: 10.1186/s12906-019-2546-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ethanol withdrawal (EtOHW) anxiety is a crucial risk factor for alcoholic relapse. The neuropeptide nociceptin/orphanin FQ (N/OFQ) acts upon its receptor (NOP) to antagonize corticotropin-releasing factor (CRF) and elicit anxiolytic actions. Semen Ziziphi Spinosae (SZS), a prototypical hypnotic-sedative herb in Oriental medicine, exhibits anxiolytic effects during nicotine withdrawal by improving amygdaloid CRF/CRF1 receptor (CRFR1) signaling. Therefore, we evaluated the effects of SZS on EtOHW anxiety and the involvement of amygdaloid CRF/CRFR1 and N/OFQ/NOP pathways. METHODS Male Sprague Dawley rats received intraperitoneal injections of 2 g/kg EtOH (20% v/v) once daily for 28 d followed by a 3-d withdrawal. During EtOHW, the rats were given once-daily intragastric treatments of a methanol extract of SZS (MESZS, 60 or 180 mg/kg/d). Anxiety-like behaviors were measured with the open field (OF) and elevated plus maze (EPM) tests, and plasma corticosterone (CORT) levels were examined by an enzyme-linked immunosorbent assay. mRNA and protein expression levels of the neuropeptides and their receptors were determined by quantitative polymerase chain reaction and Western blot assays. RESULTS MESZS increased the distance traveled in the center zone of the OF and dose-dependently elongated the duration of staying in the center zone in EtOHW rats. MESZS increased both the number of entries into and the time spent in the open arms of the EPM by EtOHW rats. And, MESZS inhibited the over secretion of plasma CORT during EtOHW. EtOHW enhanced CRF and CRFR1 gene and protein expression in the central nucleus of the amygdala (CeA), which were inhibited by 180 mg/kg/d MESZS. EtOHW increased amygdaloid NOP mRNA and protein expression but spared N/OFQ mRNA expression, and 180 mg/kg/d MESZS further promoted these increases. Additionally, a post-MESZS intra-CeA infusion of either CRF or the selective NOP antagonist UFP-101 abolished the expected anxiolytic effect of 180 mg/kg/d MESZS. CONCLUSIONS These results suggest that MESZS ameliorates EtOHW anxiety by improving both CRF/CRFR1 and N/OFQ/NOP transmissions in the CeA.
Collapse
Affiliation(s)
- Li Bo Li
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Young Woo Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Yu Hua Wang
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Li Bai
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Xiao Dong Zhu
- Department of Pharmacology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Zheng Lin Zhao
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Chul Won Lee
- Department of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Yu Jiao
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Tong Wu
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Zhen Zhen Cai
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China
| | - Sang Chan Kim
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Won G An
- Department of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Chae Ha Yang
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Guang Cheng Cui
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China.
| | - Rong Jie Zhao
- Department of Psychopharmacology, School of Mental Health, Qiqihar Medical University, 333 Bukuibei Street, Jianhua District, Qiqihar, 161006, China.
| |
Collapse
|
13
|
Logrip ML. Molecular tools to elucidate factors regulating alcohol use. Alcohol 2019; 74:3-9. [PMID: 30033149 DOI: 10.1016/j.alcohol.2018.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022]
Abstract
Alcohol use disorder (AUD) is a pervasive societal problem, marked by high levels of alcohol intake and recidivism. Despite these common disease traits, individuals diagnosed with AUD display a range of disordered drinking and alcohol-related behaviors. The diversity in disease presentation, as well as the established polygenic nature of the disorder and complex neurocircuitry, speaks to the variety of neurochemical changes resulting from alcohol intake that may differentially regulate alcohol-related behaviors. Investigations into the molecular adaptations responsible for maladaptive alcohol-related behavioral outcomes require an ever-evolving set of molecular tools to elucidate with increasing precision how alcohol alters behavior through neurochemical changes. This review highlights recent advances in molecular methodology, addressing how incorporation of these cutting-edge techniques not only may enhance current knowledge of the molecular bases of AUD, but also may facilitate identification of improved treatment targets that may be therapeutic in specific subpopulations of AUD individuals.
Collapse
|
14
|
Alsebaaly J, Dugast E, Favot L, Rabbaa Khabbaz L, Solinas M, Thiriet N. Persistent Neuroadaptations in the Expression of Genes Involved in Cholesterol Homeostasis Induced by Chronic, Voluntary Alcohol Intake in Rats. Front Mol Neurosci 2018; 11:457. [PMID: 30618609 PMCID: PMC6300585 DOI: 10.3389/fnmol.2018.00457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Alcohol use disorder (AUD) is associated with persistent adaptations in the brain that are believed to participate in the long-lasting vulnerability to relapse after abstinence. Cholesterol, the major sterol compound found in the central nervous system (CNS), plays a major role in maintenance of neuronal morphology, synaptogenesis and synaptic communication and may be involved in alcohol-induced neuroadaptations. In this study, we investigated whether alcohol consumption in a two-bottle choice paradigm followed by 3 weeks of abstinence could alter the expression of genes encoding proteins involved in cholesterol homeostasis in brain regions involved in addiction and relapse, namely the prefrontal cortex (PFC), the nucleus accumbens (NAc), the mesencephalon and the amygdala. We found that voluntary alcohol intake followed by 3 weeks of forced abstinence produces changes in the transcription of several genes encoding proteins directly involved in cholesterol synthesis such as 3-hydroxyl-3-methylglutaryl-coenzyme A (HMGCoA) reductase, farnesyl-diphosphate farnesyltransferase 1 (FDFT1) and farnesyl diphosphate synthase (FDPS) and in its regulation such as sterol regulatory element-binding factor-2 (SREBF2), in cholesterol transport such as ATP-binding cassette subfamily A member 1 (ABCA1) and in cholesterol degradation such as CYP46A1. Interestingly, these changes appeared to be region-specific and suggest that previous chronic exposure to alcohol might durably increase cholesterol metabolism in the PFC, the NAc and the mesencephalon and decrease cholesterol metabolism in the amygdala. Altogether, these results suggest that alcohol consumption leads to durable deregulations in cholesterol metabolism in key areas involved in loss of control over drug use and addiction. These long-term neuroadaptations may participate in the changes in brain structure and functioning that are responsible for the long-lasting risks of relapse to alcohol.
Collapse
Affiliation(s)
- Josette Alsebaaly
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments (LPCQM), Faculty of Pharmacy, PTS, University of Saint-Joseph of Beirut, Beirut, Lebanon
| | - Emilie Dugast
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Laure Favot
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA4331, University of Poitiers, Poitiers, France
| | - Lydia Rabbaa Khabbaz
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments (LPCQM), Faculty of Pharmacy, PTS, University of Saint-Joseph of Beirut, Beirut, Lebanon
| | - Marcello Solinas
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| | - Nathalie Thiriet
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| |
Collapse
|
15
|
CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 2018; 72:33-47. [PMID: 30217435 DOI: 10.1016/j.alcohol.2018.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 01/06/2023]
Abstract
Decades of research have described the importance of corticotropin-releasing factor (CRF) signaling in alcohol addiction, as well as in commonly co-expressed neuropsychiatric diseases, including anxiety and mood disorders. However, CRF signaling can also acutely regulate binge alcohol consumption, anxiety, and affect in non-dependent animals, possibly via modulation of central monoaminergic signaling. We hypothesize that basal CRF tone is particularly high in animals and humans with an inherent propensity for high anxiety and alcohol consumption, and thus these individuals are at increased risk for the development of alcohol use disorder and comorbid neuropsychiatric diseases. The current review focuses on extrahypothalamic CRF circuits, particularly those stemming from the bed nucleus of the stria terminalis (BNST), found to play a role in basal phenotypes, and examines whether the intrinsic hyperactivity of these circuits is sufficient to escalate the expression of these behaviors and steepen the trajectory of development of disease states. We focus our efforts on describing CRF modulation of biogenic amine neuron populations that have widespread projections to the forebrain to modulate behaviors, including alcohol and drug intake, stress reactivity, and anxiety. Further, we review the known sex differences and estradiol modulation of these neuron populations and CRF signaling at their synapses to address the question of whether females are more susceptible to the development of comorbid addiction and stress-related neuropsychiatric diseases because of hyperactive extrahypothalamic CRF circuits compared to males.
Collapse
|
16
|
Aqueous Extract of Semen Ziziphi Spinosae Exerts Anxiolytic Effects during Nicotine Withdrawal via Improvement of Amygdaloid CRF/CRF1R Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:2419183. [PMID: 30245730 PMCID: PMC6139233 DOI: 10.1155/2018/2419183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/26/2018] [Accepted: 08/16/2018] [Indexed: 12/05/2022]
Abstract
Anxiety during nicotine withdrawal (NicW) is a key risk factor for smoking relapse. Semen Ziziphi Spinosae (SZS), which is a prototypical hypnotic-sedative herb in Oriental medicine, has been clinically used to treat insomnia and general anxiety disorders for thousands of years. Thus, the present study evaluated the effects of the aqueous extract of SZS (AESZS) on NicW-induced anxiety in male rats that received subcutaneous administrations of nicotine (Nic) (0.4 mg/kg, twice a day) for 7 d followed by 4 d of withdrawal. During NicW, the rats received four intragastric treatments of AESZS (60 mg/kg/d or 180 mg/kg/d). AESZS dose-dependently attenuated NicW-induced anxiety-like behaviors in the elevated plus maze (EPM) tests and 180 mg/kg/d AESZS inhibited NicW-induced increases in plasma corticosterone. Additionally, the protein and mRNA expressions of corticotropin-releasing factor (CRF) and CRF type 1 receptor (CRF1R) increased in the central nucleus of the amygdala (CeA) during NicW, but these changes were suppressed by 180 mg/kg/d AESZS. A post-AESZS infusion of CRF into the CeA abolished the attenuation of anxiety by AESZS and 180 mg/kg/d AESZS suppressed NicW-induced increases in norepinephrine and 3-methoxy-4-hydroxy-phenylglycol levels in the CeA. The present results suggest that AESZS ameliorated NicW-induced anxiety via improvements in CRF/CRF1R and noradrenergic signaling in the CeA.
Collapse
|
17
|
Corticotropin-Releasing Factor (CRF) Neurocircuitry and Neuropharmacology in Alcohol Drinking. Handb Exp Pharmacol 2018; 248:435-471. [PMID: 29374836 DOI: 10.1007/164_2017_86] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alcohol use is pervasive in the United States. In the transition from nonhazardous drinking to hazardous drinking and alcohol use disorder, neuroadaptations occur within brain reward and brain stress systems. One brain signaling system that has received much attention in animal models of excessive alcohol drinking and alcohol dependence is corticotropin-releasing factor (CRF). The CRF system is composed of CRF, the urocortins, CRF-binding protein, and two receptors - CRF type 1 and CRF type 2. This review summarizes how acute, binge, and chronic alcohol dysregulates CRF signaling in hypothalamic and extra-hypothalamic brain regions and how this dysregulation may contribute to changes in alcohol reinforcement, excessive alcohol consumption, symptoms of negative affect during withdrawal, and alcohol relapse. In addition, it summarizes clinical work examining CRF type 1 receptor antagonists in humans and discusses why the brain CRF system is still relevant in alcohol research.
Collapse
|
18
|
Roberto M, Varodayan FP. Synaptic targets: Chronic alcohol actions. Neuropharmacology 2017; 122:85-99. [PMID: 28108359 DOI: 10.1016/j.neuropharm.2017.01.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/23/2016] [Accepted: 01/14/2017] [Indexed: 01/02/2023]
Abstract
Alcohol acts on numerous cellular and molecular targets to regulate neuronal communication within the brain. Chronic alcohol exposure and acute withdrawal generate prominent neuroadaptations at synapses, including compensatory effects on the expression, localization and function of synaptic proteins, channels and receptors. The present article reviews the literature describing the synaptic effects of chronic alcohol exposure and their relevance for synaptic transmission in the central nervous system. This review is not meant to be comprehensive, but rather to highlight the effects that have been observed most consistently and that are thought to contribute to the development of alcohol dependence and the negative aspects of withdrawal. Specifically, we will focus on the major excitatory and inhibitory neurotransmitters in the brain, glutamate and GABA, respectively, and how their neuroadaptations after chronic alcohol exposure contributes to alcohol reinforcement, dependence and withdrawal. This article is part of the Special Issue entitled "Alcoholism".
Collapse
|
19
|
Quadros IMH, Macedo GC, Domingues LP, Favoretto CA. An Update on CRF Mechanisms Underlying Alcohol Use Disorders and Dependence. Front Endocrinol (Lausanne) 2016; 7:134. [PMID: 27818644 PMCID: PMC5073134 DOI: 10.3389/fendo.2016.00134] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022] Open
Abstract
Alcohol is the most commonly used and abused substance worldwide. The emergence of alcohol use disorders, and alcohol dependence in particular, is accompanied by functional changes in brain reward and stress systems, which contribute to escalated alcohol drinking and seeking. Corticotropin-releasing factor (CRF) systems have been critically implied in the transition toward problematic alcohol drinking and alcohol dependence. This review will discuss how dysregulation of CRF function contributes to the vulnerability for escalated alcohol drinking and other consequences of alcohol consumption, based on preclinical evidence. CRF signaling, mostly via CRF1 receptors, seems to be particularly important in conditions of excessive alcohol taking and seeking, including during early and protracted withdrawal, relapse, as well as during withdrawal-induced anxiety and escalated aggression promoted by alcohol. Modulation of CRF1 function seems to exert a less prominent role over low to moderate alcohol intake, or to species-typical behaviors. While CRF mechanisms in the hypothalamic-pituitary-adrenal axis have some contribution to the neurobiology of alcohol abuse and dependence, a pivotal role for extra-hypothalamic CRF pathways, particularly in the extended amygdala, is well characterized. More recent studies further suggest a direct modulation of brain reward function by CRF signaling in the ventral tegmental area, nucleus accumbens, and the prefrontal cortex, among other structures. This review will further discuss a putative role for other components of the CRF system that contribute for the overall balance of CRF function in reward and stress pathways, including CRF2 receptors, CRF-binding protein, and urocortins, a family of CRF-related peptides.
Collapse
Affiliation(s)
- Isabel Marian Hartmann Quadros
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Giovana Camila Macedo
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Liz Paola Domingues
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane Aparecida Favoretto
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Ketchesin KD, Stinnett GS, Seasholtz AF. Binge Drinking Decreases Corticotropin-Releasing Factor-Binding Protein Expression in the Medial Prefrontal Cortex of Mice. Alcohol Clin Exp Res 2016; 40:1641-50. [PMID: 27374820 DOI: 10.1111/acer.13119] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/29/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND Dysregulation of the corticotropin-releasing factor (CRF) system has been observed in rodent models of binge drinking, with a large focus on CRF receptor 1 (CRF-R1). The role of CRF-binding protein (CRF-BP), a key regulator of CRF activity, in binge drinking is less well understood. In humans, single-nucleotide polymorphisms in CRHBP are associated with alcohol use disorder and stress-induced alcohol craving, suggesting a role for CRF-BP in vulnerability to alcohol addiction. METHODS The role and regulation of CRF-BP in binge drinking were examined in mice exposed to the drinking in the dark (DID) paradigm. Using in situ hybridization, the regulation of CRF-BP, CRF-R1, and CRF mRNA expression was determined in the stress and reward systems of C57BL/6J mice after repeated cycles of DID. To determine the functional role of CRF-BP in binge drinking, CRF-BP knockout (CRF-BP KO) mice were exposed to 6 cycles of DID, during which alcohol consumption was measured and compared to wild-type mice. RESULTS CRF-BP mRNA expression was significantly decreased in the prelimbic (PL) and infralimbic medial prefrontal cortex (mPFC) of C57BL/6J mice after 3 cycles and in the PL mPFC after 6 cycles of DID. No significant changes in CRF or CRF-R1 mRNA levels were observed in mPFC, ventral tegmental area, bed nucleus of the stria terminalis, or amygdala after 3 cycles of DID. CRF-BP KO mice do not show significant alterations in drinking compared to wild-type mice across 6 cycles of DID. CONCLUSIONS These results reveal that repeated cycles of binge drinking alter CRF-BP mRNA expression in the mPFC, a region responsible for executive function and regulation of emotion and behavior, including responses to stress. We observed a persistent decrease in CRF-BP mRNA expression in the mPFC after 3 and 6 DID cycles, which may allow for increased CRF signaling at CRF-R1 and contribute to excessive binge-like ethanol consumption.
Collapse
Affiliation(s)
- Kyle D Ketchesin
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Gwen S Stinnett
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Audrey F Seasholtz
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan.,Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
21
|
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: 38] [Impact Index Per Article: 3.8] [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
|
22
|
Barker JM, Taylor JR. Habitual alcohol seeking: modeling the transition from casual drinking to addiction. Neurosci Biobehav Rev 2014; 47:281-94. [PMID: 25193245 PMCID: PMC4258136 DOI: 10.1016/j.neubiorev.2014.08.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/17/2014] [Accepted: 08/25/2014] [Indexed: 12/29/2022]
Abstract
The transition from goal-directed actions to habitual ethanol seeking models the development of addictive behavior that characterizes alcohol use disorders. The progression to habitual ethanol-seeking behavior occurs more rapidly than for natural rewards, suggesting that ethanol may act on habit circuit to drive the loss of behavioral flexibility. This review will highlight recent research that has focused on the formation and expression of habitual ethanol seeking, and the commonalities and distinctions between ethanol and natural reward-seeking habits, with the goal of highlighting important, understudied research areas that we believe will lead toward the development of novel treatment and prevention strategies for uncontrolled drinking.
Collapse
Affiliation(s)
- Jacqueline M Barker
- Department of Psychiatry, Yale University School of Medicine, Ribicoff Labs, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jane R Taylor
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
23
|
Jin Z, Bhandage AK, Bazov I, Kononenko O, Bakalkin G, Korpi ER, Birnir B. Expression of specific ionotropic glutamate and GABA-A receptor subunits is decreased in central amygdala of alcoholics. Front Cell Neurosci 2014; 8:288. [PMID: 25278838 PMCID: PMC4165314 DOI: 10.3389/fncel.2014.00288] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/29/2014] [Indexed: 01/20/2023] Open
Abstract
The central amygdala (CeA) has a role for mediating fear and anxiety responses. It is also involved in emotional imbalance caused by alcohol abuse and dependence and in regulating relapse to alcohol abuse. Growing evidences suggest that excitatory glutamatergic and inhibitory γ-aminobutyric acid-ergic (GABAergic) transmissions in the CeA are affected by chronic alcohol exposure. Human post-mortem CeA samples from male alcoholics (n = 9) and matched controls (n = 9) were assayed for the expression level of ionotropic glutamate and GABA-A receptors subunit mRNAs using quantitative real-time reverse transcription-PCR (RT-qPCR). Our data revealed that out of the 16 ionotropic glutamate receptor subunits, mRNAs encoding two AMPA [2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid] receptor subunits GluA1 and GluA4; one kainate receptor subunit GluK2; one NMDA (N-methyl-D-aspartate) receptor subunit GluN2D and one delta receptor subunit GluD2 were significantly decreased in the CeA of alcoholics. In contrast, of the 19 GABA-A receptor subunits, only the mRNA encoding the α2 subunit was significantly down-regulated in the CeA of the alcoholics as compared with control subjects. Our findings imply that the down-regulation of specific ionotropic glutamate and GABA-A receptor subunits in the CeA of alcoholics may represent one of the molecular substrates underlying the new balance between excitatory and inhibitory neurotransmission in alcohol dependence.
Collapse
Affiliation(s)
- Zhe Jin
- Molecular Physiology and Neuroscience Unit, Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| | - Amol K Bhandage
- Molecular Physiology and Neuroscience Unit, Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| | - Igor Bazov
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Olga Kononenko
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Georgy Bakalkin
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Esa R Korpi
- Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland
| | - Bryndis Birnir
- Molecular Physiology and Neuroscience Unit, Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| |
Collapse
|
24
|
Xia X, Liu R, Li Y, Xue S, Liu Q, Jiang X, Zhang W, Ding K. Cloning and molecular characterization of scorpion Buthus martensi venom hyaluronidases: a novel full-length and diversiform noncoding isoforms. Gene 2014; 547:338-45. [PMID: 24973698 DOI: 10.1016/j.gene.2014.06.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/06/2014] [Accepted: 06/23/2014] [Indexed: 11/16/2022]
Abstract
Hyaluronidase is a common component of scorpion venom and has been considered as "spreading factor" that promotes a fast penetration of the venom in the anaphylactic reaction. In the current study, a novel full-length of hyaluronidase BmHYI and three noncoding isoforms of BmHYII, BmHYIII and BmHYIV were cloned by using a combined strategy based on peptide sequencing and Rapid Amplification of cDNA Ends (RACE). BmHYI has 410 amino acid residues containing the catalytic, positional and five potential N-glycosylation sites. The deduced protein sequence of BmHYI shares significant identity with venom hyaluronidases from bees and snakes. The phylogenetic analysis showed early divergence and independent evolution of BmHYI from other hyaluronidases. An extraordinarily high level of sequence similarity was detected among four sequences. But, BmHYII, BmHYIII and BmHYIV were short of stop-codon in the open reading frame and poly(A) signal in the 3' end.
Collapse
Affiliation(s)
- Xichao Xia
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China.
| | - Rongzhi Liu
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Yi Li
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Shipeng Xue
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Qingchun Liu
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Xiao Jiang
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Wenjuan Zhang
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| | - Ke Ding
- Basal Medical College of Nanyang Medical University, Nanyang, 473041 Henan Province, China
| |
Collapse
|
25
|
Zamora-Martinez ER, Edwards S. Neuronal extracellular signal-regulated kinase (ERK) activity as marker and mediator of alcohol and opioid dependence. Front Integr Neurosci 2014; 8:24. [PMID: 24653683 PMCID: PMC3949304 DOI: 10.3389/fnint.2014.00024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/19/2014] [Indexed: 11/13/2022] Open
Abstract
Early pioneering work in the field of biochemistry identified phosphorylation as a crucial post-translational modification of proteins with the ability to both indicate and arbitrate complex physiological processes. More recent investigations have functionally linked phosphorylation of extracellular signal-regulated kinase (ERK) to a variety of neurophysiological mechanisms ranging from acute neurotransmitter action to long-term gene expression. ERK phosphorylation serves as an intracellular bridging mechanism that facilitates neuronal communication and plasticity. Drugs of abuse, including alcohol and opioids, act as artificial yet powerful rewards that impinge upon natural reinforcement processes critical for survival. The graded progression from initial exposure to addiction (or substance dependence) is believed to result from drug- and drug context-induced adaptations in neuronal signaling processes across brain reward and stress circuits following excessive drug use. In this regard, commonly abused drugs as well as drug-associated experiences are capable of modifying the phosphorylation of ERK within central reinforcement systems. In addition, chronic drug and alcohol exposure may drive ERK-regulated epigenetic and structural alterations that underlie a long-term propensity for escalating drug use. Under the influence of such a neurobiological vulnerability, encountering drug-associated cues and contexts can produce subsequent alterations in ERK signaling that drive relapse to drug and alcohol seeking. Current studies are determining precisely which molecular and regional ERK phosphorylation-associated events contribute to the addiction process, as well as which neuroadaptations need to be targeted in order to return dependent individuals to a healthy state.
Collapse
Affiliation(s)
- Eva R Zamora-Martinez
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute La Jolla, CA, USA
| | - Scott Edwards
- Department of Physiology and Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
26
|
Lindsay JH, Glass JD, Amicarelli M, Prosser RA. The mammalian circadian clock in the suprachiasmatic nucleus exhibits rapid tolerance to ethanol in vivo and in vitro. Alcohol Clin Exp Res 2014; 38:760-9. [PMID: 24512529 DOI: 10.1111/acer.12303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/16/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ethanol (EtOH) triggers cellular adaptations that induce tolerance in many brain areas, including the suprachiasmatic nucleus (SCN), the site of the master circadian clock. EtOH inhibits light-induced phase shifts in the SCN in vivo and glutamate-induced phase shifts in vitro. The in vitro phase shifts develop acute tolerance to EtOH, occurring within minutes of initial exposure, while the in vivo phase shifts exhibit no evidence of chronic tolerance. An intermediate form, rapid tolerance, is not well studied but may predict subsequent chronic tolerance. Here, we investigated rapid tolerance in the SCN clock. METHODS Adult C57BL/6 mice were provided 15% EtOH or water for one 12-hour lights-off period. For in vitro experiments, SCN-containing brain slices were prepared in the morning and treated for 10 minutes with glutamate +/- EtOH the following night. Single-cell neuronal firing rates were recorded extracellularly during the subsequent day to determine SCN clock phase. For in vivo experiments, mice receiving EtOH 24 hours previously were exposed to a 30-minute light pulse immediately preceded by intraperitoneal saline or 2 g/kg EtOH injection. Mice were then placed in constant darkness and their phase-shifting responses measured. RESULTS In vitro, the SCN clock from EtOH-exposed mice exhibited rapid tolerance, with a 10-fold increase in EtOH needed to inhibit glutamate-induced phase shifts. Co-application of brain-derived neurotrophic factor prevented EtOH inhibition, consistent with experiments using EtOH-naïve mice. Rapid tolerance lasts 48 to 96 hours, depending on whether assessing in vitro phase advances or phase delays. Similarly, in vivo, prior EtOH consumption prevented EtOH's acute blockade of photic phase delays. Finally, immunoblot experiments showed no changes in SCN glutamate receptor subunit (NR2B) expression or phosphorylation in response to rapid tolerance induction. CONCLUSIONS The SCN circadian clock develops rapid tolerance to EtOH as assessed both in vivo and in vitro, and the tolerance lasts for several days. These data demonstrate the utility of the circadian system as a model for investigating cellular mechanisms through which EtOH acts in the brain.
Collapse
Affiliation(s)
- Jonathan H Lindsay
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | | | | | | |
Collapse
|
27
|
Jonsson S, Ericson M, Söderpalm B. Modest Long-Term Ethanol Consumption Affects Expression of Neurotransmitter Receptor Genes in the Rat Nucleus Accumbens. Alcohol Clin Exp Res 2013; 38:722-9. [DOI: 10.1111/acer.12307] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 09/27/2013] [Indexed: 01/21/2023]
Affiliation(s)
- Susanne Jonsson
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg and Beroendekliniken; Sahlgrenska University Hospital; Gothenburg Sweden
| | - Mia Ericson
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg and Beroendekliniken; Sahlgrenska University Hospital; Gothenburg Sweden
| | - Bo Söderpalm
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg and Beroendekliniken; Sahlgrenska University Hospital; Gothenburg Sweden
| |
Collapse
|
28
|
Abstract
RATIONALE An increasingly compelling literature points to a major role for the glutamate system in mediating the effects of alcohol on behavior and the pathophysiology of alcoholism. Preclinical studies indicate that glutamate signaling mediates certain aspects of ethanol's intoxicating and rewarding effects, and undergoes adaptations following chronic alcohol exposure that may contribute to the withdrawal, craving and compulsive drug-seeking that drive alcohol abuse and alcoholism. OBJECTIVES We discuss the potential for targeting the glutamate system as a novel pharmacotherapeutic approach to treating alcohol use disorders, focusing on five major components of the glutamate system: the N-methyl-D-aspartate (NMDA) receptor and specific NMDA subunits, the glycineB site on the NMDA receptors (NMDAR), L-alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid ionotropic (AMPA) and kainate (KAR) receptors, metabotropic receptors (mGluR), and glutamate transporters. RESULTS Chronic alcohol abuse produces a hyperglutamatergic state, characterized by elevated extracellular glutamate and altered glutamate receptors and transporters. Pharmacologically manipulating glutamatergic neurotransmission alters alcohol-related behaviors including intoxication, withdrawal, and alcohol-seeking, in rodents and human subjects. Blocking NMDA and AMPA receptors reduces alcohol consumption in rodents, but side-effects may limit this as a therapeutic approach. Selectively targeting NMDA and AMPA receptor subunits (e.g., GluN2B, GluA3), or the NMDAR glycineB site offers an alternative approach. Blocking mGluR5 potently affects various alcohol-related behaviors in rodents, and mGluR2/3 agonism also suppresses alcohol consumption. Finally, glutamate transporter upregulation may mitigate behavioral and neurotoxic sequelae of excess glutamate caused by alcohol. CONCLUSIONS Despite the many challenges that remain, targeting the glutamate system offers genuine promise for developing new treatments for alcoholism.
Collapse
|
29
|
Effects of fluoxetine on CRF and CRF1 expression in rats exposed to the learned helplessness paradigm. Psychopharmacology (Berl) 2013; 225:647-59. [PMID: 22960774 DOI: 10.1007/s00213-012-2859-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 08/16/2012] [Indexed: 12/25/2022]
Abstract
RATIONALE Stress is a common antecedent reported by people suffering major depression. In these patients, extrahypothalamic brain areas, like the hippocampus and basolateral amygdala (BLA), have been found to be affected. The BLA synthesizes CRF, a mediator of the stress response, and projects to hippocampus. The main hippocampal target for this peptide is the CRF subtype 1 receptor (CRF1). Evidence points to a relationship between dysregulation of CRF/CRF1 extrahypothalamic signaling and depression. OBJECTIVE Because selective serotonin reuptake inhibitors (SSRIs) are the first-line pharmacological treatment for depression, we investigated the effect of chronic treatment with the SSRI fluoxetine on long-term changes in CRF/CRF1 signaling in animals showing a depressive-like behavior. METHODS Male Wistar rats were exposed to the learned helplessness paradigm (LH). After evaluation of behavioral impairment, the animals were treated with fluoxetine (10 mg/kg i.p.) or saline for 21 days. We measured BLA CRF expression with RT-PCR and CRF1 expression in CA3 and the dentate gyrus of the hippocampus with in situ hybridization. We also studied the activation of one of CRF1's major intracellular signaling targets, the extracellular signal-related kinases 1 and 2 (ERK1/2) in CA3. RESULTS In saline-treated LH animals, CRF expression in the BLA increased, while hippocampal CRF1 expression and ERK1/2 activation decreased. Treatment with fluoxetine reversed the changes in CRF and CRF1 expressions, but not in ERK1/2 activation. CONCLUSION In animals exposed to the learned helplessness paradigm, there are long-term changes in CRF and CRF1 expression that are restored with a behaviorally effective antidepressant treatment.
Collapse
|
30
|
Roberto M, Gilpin NW, Siggins GR. The central amygdala and alcohol: role of γ-aminobutyric acid, glutamate, and neuropeptides. Cold Spring Harb Perspect Med 2012; 2:a012195. [PMID: 23085848 PMCID: PMC3543070 DOI: 10.1101/cshperspect.a012195] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug seeking and drug taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptative mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid-ergic (GABAergic) transmission in CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and AMPA receptors in CeA, whereas chronic alcohol up-regulates N-methyl-d-aspartate receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and anti-stress (e.g., NPY, nociceptin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
31
|
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
|
32
|
Haass-Koffler CL, Bartlett SE. Stress and addiction: contribution of the corticotropin releasing factor (CRF) system in neuroplasticity. Front Mol Neurosci 2012; 5:91. [PMID: 22973190 PMCID: PMC3434418 DOI: 10.3389/fnmol.2012.00091] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/15/2012] [Indexed: 12/22/2022] Open
Abstract
Corticotropin releasing factor (CRF) has been shown to induce various behavioral changes related to adaptation to stress. Dysregulation of the CRF system at any point can lead to a variety of psychiatric disorders, including substance use disorders (SUDs). CRF has been associated with stress-induced drug reinforcement. Extensive literature has identified CRF to play an important role in the molecular mechanisms that lead to an increase in susceptibility that precipitates relapse to SUDs. The CRF system has a heterogeneous role in SUDs. It enhances the acute effects of drugs of abuse and is also responsible for the potentiation of drug-induced neuroplasticity evoked during the withdrawal period. We present in this review the brain regions and circuitries where CRF is expressed and may participate in stress-induced drug abuse. Finally, we attempt to evaluate the role of modulating the CRF system as a possible therapeutic strategy for treating the dysregulation of emotional behaviors that result from the acute positive reinforcement of substances of abuse as well as the negative reinforcement produced by withdrawal.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Ernest Gallo Clinic and Research Center at the University of California San Francisco Emeryville, CA, USA
| | | |
Collapse
|
33
|
Corticotropin releasing factor signaling in the central amygdala is recruited during binge-like ethanol consumption in C57BL/6J mice. J Neurosci 2012; 32:3405-13. [PMID: 22399763 DOI: 10.1523/jneurosci.6256-11.2012] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A well established body of work indicates a crucial role for corticotropin-releasing factor (CRF) in neurobiological responses associated with excessive dependence-like ethanol drinking in ethanol-vapor-exposed rodents. Recent evidence demonstrates a role for CRF in the modulation of binge-like ethanol consumption by nondependent mice, a behavior that can precede ethanol dependence. The CRF circuitry that is engaged by binge-like ethanol exposure, however, is unknown. Using converging approaches, we provide evidence that, similar to ethanol-vapor-induced increases in ethanol intake, CRF signaling in the central nucleus of the amygdala (CeA) is engaged during binge-like ethanol consumption by C57BL/6J mice. Specifically, we found that binge-like consumption of an ethanol solution (20% ethanol v/v) was attenuated by pretreatment with the CRF1R antagonists antalarmin, 4-ethyl-[2,5,6-trimethyl-7-(2,4,6-trimethylphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]amino-1-butanol, and NBI-27914 at doses (30 mg/kg, i.p.) that did not alter nonbinge-like ethanol consumption. Binge-like ethanol consumption resulted in significant increases of CRF immunoreactivity in the CeA immediately following ethanol drinking and 18-24 h following ethanol removal and also blocked the ability of CRF to enhance GABAergic transmission in the CeA 18-24 h following ethanol removal. Pretreatment with bilateral injections of antalarmin (1 μg/0.5 μl per side) into the CeA, but not the adjacent basolateral amygdala, significantly attenuated binge-like ethanol consumption. These findings suggest that CRF signaling in the CeA is recruited during excessive ethanol intake, before the development of dependence. We hypothesize that plastic changes in CRF signaling develop with repeated binge-like drinking episodes, contributing to the transition to dependence.
Collapse
|
34
|
Prendergast MA, Mulholland PJ. Glucocorticoid and polyamine interactions in the plasticity of glutamatergic synapses that contribute to ethanol-associated dependence and neuronal injury. Addict Biol 2012; 17:209-23. [PMID: 21967628 PMCID: PMC3254017 DOI: 10.1111/j.1369-1600.2011.00375.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Stress contributes to the development of ethanol dependence and is also a consequence of dependence. However, the complexity of physiological interactions between activation of the hypothalamic-pituitary-adrenal (HPA) axis and ethanol itself is not well delineated. Emerging evidence derived from examination of corticotropin-releasing factor systems and glucocorticoid receptor systems in ethanol dependence suggests a role for pharmacological manipulation of the HPA axis in attenuating ethanol intake, though it is not clear how activation of the HPA axis may promote ethanol dependence or contribute to the neuroadaptative changes that accompany the development of dependence and the severity of ethanol withdrawal. This review examines the role that glucocorticoids, in particular, have in promoting ethanol-associated plasticity of glutamatergic synapses by influencing expression of endogenous linear polyamines and polyamine-sensitive polypeptide subunits of N-methyl-D-aspartate (NMDA)-type glutamate receptors. We provide evidence that interactions among glucocorticoid systems, polyamines and NMDA receptors are highly relevant to both the development of ethanol dependence and to behavioral and neuropathological sequelae associated with ethanol withdrawal. Examination of these issues is likely to be of critical importance not only in further elucidating the neurobiology of HPA axis dysregulation in ethanol dependence, but also with regard to identification of novel therapeutic targets that may be exploited in the treatment of ethanol dependence.
Collapse
Affiliation(s)
- Mark A. Prendergast
- University of Kentucky, Department of Psychology, 741 South Limestone Street, Lexington, KY 40536, U.S.A
- Spinal Cord and Brain Injury Research Center, B449 Biomedical and Biological Sciences Research Building, 741 South Limestone Street, Lexington, KY 40536, U.S.A
| | - Patrick J. Mulholland
- Departments of Neurosciences and Psychiatry & Behavioral Sciences, Medical University of South Carolina, 67 President Street, IOP 462 North Charleston, South Carolina 29425, U.S.A
| |
Collapse
|
35
|
Christian DT, Alexander NJ, Diaz MR, Robinson S, McCool BA. Chronic intermittent ethanol and withdrawal differentially modulate basolateral amygdala AMPA-type glutamate receptor function and trafficking. Neuropharmacology 2012; 62:2430-9. [PMID: 22387532 DOI: 10.1016/j.neuropharm.2012.02.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/15/2012] [Accepted: 02/16/2012] [Indexed: 12/19/2022]
Abstract
The amygdala plays a critical role in the generation and expression of anxiety-like behaviors including those expressed following withdrawal (WD) from chronic intermittent ethanol (CIE) exposure. In particular, the BLA glutamatergic system controls the expression of both innate and pathological anxiety. Recent data suggests that CIE and WD may functionally alter this system in a manner that closely parallels memory-related phenomena like long-term potentiation (LTP). We therefore specifically dissected CIE/WD-induced changes in glutamatergic signaling using electrophysiological and biochemical approaches with a particular focus on the plasticity-related components of this neurotransmitter system. Our results indicate that cortical glutamatergic inputs arriving at BLA principal via the external capsule undergo predominantly post-synaptic alterations in AMPA receptor function following CIE and WD. Biochemical analysis revealed treatment-dependent changes in AMPA receptor surface expression and subunit phosphorylation that are complemented by changes in total protein levels and/or phosphorylation status of several key, plasticity-associated protein kinases such as calcium/calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC). Together, these data show that CIE- and WD-induced changes in BLA glutamatergic function both functionally and biochemically mimic plasticity-related states. These mechanisms likely contribute to long-term increases in anxiety-like behavior following chronic ethanol exposure.
Collapse
Affiliation(s)
- Daniel T Christian
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | | |
Collapse
|
36
|
Gilpin NW, Roberto M. Neuropeptide modulation of central amygdala neuroplasticity is a key mediator of alcohol dependence. Neurosci Biobehav Rev 2012; 36:873-88. [PMID: 22101113 PMCID: PMC3325612 DOI: 10.1016/j.neubiorev.2011.11.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 11/01/2011] [Accepted: 11/05/2011] [Indexed: 01/08/2023]
Abstract
Alcohol use disorders are characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and withdrawal syndrome in the absence of drug. The central amygdala (CeA) and neighboring regions (extended amygdala) mediate alcohol-related behaviors and chronic alcohol-induced plasticity. Acute alcohol suppresses excitatory (glutamatergic) transmission whereas chronic alcohol enhances glutamatergic transmission in CeA. Acute alcohol facilitates inhibitory (GABAergic) transmission in CeA, and chronic alcohol increases GABAergic transmission. Electrophysiology techniques are used to explore the effects of neuropeptides/neuromodulators (CRF, NPY, nociceptin, dynorphin, endocannabinoids, galanin) on inhibitory transmission in CeA. In general, pro-anxiety peptides increase, and anti-anxiety peptides decrease CeA GABAergic transmission. These neuropeptides facilitate or block the action of acute alcohol in CeA, and chronic alcohol produces plasticity in neuropeptide systems, possibly reflecting recruitment of negative reinforcement mechanisms during the transition to alcohol dependence. A disinhibition model of CeA output is discussed in the context of alcohol dependence- and anxiety-related behaviors.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA.
| | | |
Collapse
|
37
|
Edwards S, Guerrero M, Ghoneim OM, Roberts E, Koob GF. Evidence that vasopressin V1b receptors mediate the transition to excessive drinking in ethanol-dependent rats. Addict Biol 2012; 17:76-85. [PMID: 21309953 DOI: 10.1111/j.1369-1600.2010.00291.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alcoholism is a devastating condition that represents a progression from initial alcohol use to dependence. Although most individuals are capable of consuming alcohol in a limited fashion, the development of alcohol dependence in a subset of individuals is often associated with negative emotional states (including anxiety and depression). Since the alleviation of this negative motivational state via excessive alcohol consumption often becomes a central goal of alcoholics, the transition from initial use to dependence is postulated to be associated with a transition from positive to negative reinforcement mechanisms. Vasopressin is a neuropeptide known to potentiate the effects of CRF on the HPA axis, and emerging evidence also suggests a role for centrally located vasopressin acting on V(1b) receptors in the regulation of stress- and anxiety-like behaviors in rodents. The present study determined state-dependent alterations in vasopressin/V(1b) R signaling in an animal model of ethanol dependence. The V(1b) R antagonist SSR149415 dose-dependently reduced excessive levels of ethanol self-administration observed in dependent animals without affecting the limited levels of ethanol drinking in non-dependent animals. Ethanol self-administration reduced V(1b) receptor levels in the basolateral amygdala of non-dependent animals, a neuroadaptation that could theoretically facilitate the positive reinforcing effects of alcohol. In contrast, V(1b) R levels were seemingly restored in ethanol-dependent rats, a switch that may in part underlie a transition from positive to negative reinforcement mechanisms with dependence. Together, our data suggest a key role for vasopressin/V(1b) R signaling in the transition to ethanol dependence.
Collapse
Affiliation(s)
- Scott Edwards
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
38
|
Lam MP, Gianoulakis C. Effects of corticotropin-releasing hormone receptor antagonists on the ethanol-induced increase of dynorphin A1-8 release in the rat central amygdala. Alcohol 2011; 45:621-30. [PMID: 21840158 DOI: 10.1016/j.alcohol.2011.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 05/24/2011] [Accepted: 05/24/2011] [Indexed: 02/02/2023]
Abstract
Neurons in the central amygdala (CeA) co-express dynorphin and corticotropin-releasing hormone (CRH). Moreover, the activity of both the CRH and dynorphin systems in CeA is altered by alcohol treatments, effects suggesting interactions between the CRH and dynorphin systems. Thus, the objectives of the present study were to investigate the effects of (1) activating CRH receptors (CRHRs) by microinjection of CRH in CeA and (2) blocking CRHRs by local microinjections of CRHR antagonists in the CeA on the alcohol-induced changes in the extracellular concentrations of dynorphin A1-8 with in vivo microdialysis experiments. Microdialysis probes with a microinjection port were implanted in the CeA of alcohol-naïve Sprague-Dawley rats. Microinjections of CRH or antalarmin, a CRH receptor type 1 (CRHR1) antagonist, or anti-sauvagine-30, a CRH receptor type 2 (CRHR2) antagonist, at the level of CeA were followed by an intraperitoneal injection of either saline or 2.8 g ethanol/kg body weight. The content of dynorphin A1-8 was determined in dialyzate samples obtained prior to and following the various treatments using a specific radioimmunoassay. Activation of CRHRs in CeA induced an increase in the extracellular concentrations of dynorphin A1-8. Moreover, acute alcohol administration increased the extracellular concentrations of dynorphin A1-8 in CeA, an effect that was attenuated by blocking CRHR2 with anti-sauvagine-30 microinjection but not blocking CRHR1 with antalarmin microinjection. Therefore, the findings suggest an interaction between the CRH and dynorphin A1-8 systems at the level of CeA in response to acute alcohol exposure.
Collapse
|
39
|
McCool BA, Christian DT, Diaz MR, Läck AK. Glutamate plasticity in the drunken amygdala: the making of an anxious synapse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 91:205-33. [PMID: 20813244 DOI: 10.1016/s0074-7742(10)91007-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Plasticity at glutamatergic synapses is believed to be the cellular correlate of learning and memory. Classic fear conditioning, for example, is dependent upon NMDA-type glutamate receptor activation in the lateral/basolateral amygdala followed by increased synaptic expression of AMPA-type glutamate receptors. This review provides an extensive comparison between the initiation and expression of glutamatergic plasticity during learning/memory and glutamatergic alterations associated with chronic ethanol exposure and withdrawal. The parallels between these neuro-adaptive processes suggest that long-term ethanol exposure might "chemically condition" amygdala-dependent fear/anxiety via the increased function of pre- and post-synaptic glutamate signaling.
Collapse
Affiliation(s)
- Brian A McCool
- Department of Physiology & Pharmcology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
40
|
Femenía T, García-Gutiérrez MS, Manzanares J. CB1 receptor blockade decreases ethanol intake and associated neurochemical changes in fawn-hooded rats. Alcohol Clin Exp Res 2009; 34:131-41. [PMID: 19860799 DOI: 10.1111/j.1530-0277.2009.01074.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND This study was undertaken to identify the neurochemical changes underlying the attenuation of voluntary ethanol intake induced by the cannabinoid CB1 receptor antagonist AM251 in fawn-hooded rats. METHODS Rats were exposed to the 2-bottle-choice paradigm (ethanol 10% v/v or water) for 15 days. After this period, rats received AM251 (3 to 6 mg/kg, i.p.) or vehicle. RESULTS Voluntary ethanol intake decreased (30%) with the administration of incremental dosages of AM251 (3 mg/kg, 5 days and 6 mg/kg, 5 days) in rats with acquired high preferring ethanol consumption (>3.5 g of ethanol/kg/d). Ethanol intake significantly decreased proopiomelanocortin expression in the arcuate nucleus (38.31%) and micro-opioid-DAMGO-stimulated [(35)S]-GTPgamma binding in the caudate-putamen (40%), nucleus accumbens core (AccC) (32.87%), and shell (AccS) (34.21%). Moreover, ethanol intake increased tyrosine hydroxylase (TH) gene expression in the substantia nigra (24%) and ventral tegmental area (23%) and corticotrophin-releasing gene expression in the paraventricular hypothalamic nucleus (41.6%). The reduction of ethanol intake induced by AM251 was associated with blockade or significant reduction of the changes produced by ethanol in the expression of these genes in key regions related to drug dependence. Interestingly, treatment with AM251 reduced (20%) TH gene expression in rats drinking only water. In this respect, the action of AM251 in reducing TH gene expression may not be specific. CONCLUSION Taken together, these results revealed that blockade of cannabinoid CB1 receptors (CB1r) decreased voluntary ethanol intake in ethanol-habituated rats by normalizing the neurochemical alterations induced by ethanol.
Collapse
Affiliation(s)
- Teresa Femenía
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | | | |
Collapse
|
41
|
Bell RL, Kimpel MW, McClintick JN, Strother WN, Carr LG, Liang T, Rodd ZA, Mayfield RD, Edenberg HJ, McBride WJ. Gene expression changes in the nucleus accumbens of alcohol-preferring rats following chronic ethanol consumption. Pharmacol Biochem Behav 2009; 94:131-47. [PMID: 19666046 DOI: 10.1016/j.pbb.2009.07.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 07/21/2009] [Accepted: 07/28/2009] [Indexed: 12/16/2022]
Abstract
The objective of this study was to determine the effects of binge-like alcohol drinking on gene expression changes in the nucleus accumbens (ACB) of alcohol-preferring (P) rats. Adult male P rats were given ethanol under multiple scheduled access (MSA; three 1-h dark cycle sessions/day) conditions for 8 weeks. For comparison purposes, a second ethanol drinking group was given continuous/daily alcohol access (CA; 24h/day). A third group was ethanol-naïve (W group). Average ethanol intakes for the CA and MSA groups were approximately 9.5 and 6.5 g/kg/day, respectively. Fifteen hours after the last drinking episode, rats were euthanized, the brains extracted, and the ACB dissected. RNA was extracted and purified for microarray analysis. The only significant differences were between the CA and W groups (p<0.01; Storey false discovery rate=0.15); there were 374 differences in named genes between these 2 groups. There were 20 significant Gene Ontology (GO) categories, which included negative regulation of protein kinase activity, anti-apoptosis, and regulation of G-protein coupled receptor signaling. Ingenuity analysis indicated a network of transcription factors, involving oncogenes (Fos, Jun, Junb had higher expression in the ACB of the CA group), suggesting increased neuronal activity. There were 43 genes located within rat QTLs for alcohol consumption and preference; 4 of these genes (Tgfa, Hspa5, Mtus1 and Creb3l2) are involved in anti-apoptosis and increased transcription, suggesting that they may be contributing to cellular protection and maintaining high alcohol intakes. Overall, these findings suggest that chronic CA drinking results in genomic changes that can be observed during the early acute phase of ethanol withdrawal. Conversely, chronic MSA drinking, with its associated protracted withdrawal periods, results in genomic changes that may be masked by tight regulation of these genes following repeated experiences of ethanol withdrawal.
Collapse
Affiliation(s)
- Richard L Bell
- Department of Psychiatry, Indiana University Purdue University at Indianapolis, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Persisting changes in basolateral amygdala mRNAs after chronic ethanol consumption. Physiol Behav 2009; 96:169-73. [DOI: 10.1016/j.physbeh.2008.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 08/29/2008] [Accepted: 09/25/2008] [Indexed: 11/19/2022]
|
43
|
Henson MA, Roberts AC, Salimi K, Vadlamudi S, Hamer RM, Gilmore JH, Jarskog LF, Philpot BD. Developmental regulation of the NMDA receptor subunits, NR3A and NR1, in human prefrontal cortex. Cereb Cortex 2008; 18:2560-73. [PMID: 18296432 PMCID: PMC2733318 DOI: 10.1093/cercor/bhn017] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Subunit composition of N-methyl-D-aspartate-type glutamate receptors (NMDARs) dictates their function, yet the ontogenic profiles of human NMDAR subunits from gestation to adulthood have not been determined. We examined NMDAR mRNA and protein development in human dorsolateral prefrontal cortex (DLPFC), an area in which NMDARs are critical for higher cognitive processing and NMDAR hypofunction is hypothesized in schizophrenia. Using quantitative reverse transcriptase-polymerase chain reaction and western blotting, we found NR1 expression begins low prenatally, peaks in adolescence, yet remains high throughout life, suggesting lifelong importance of NMDAR function. In contrast, NR3A levels are low during gestation, surge soon after birth, and decline progressively through adolescence and into adulthood. Because NR3A subunits uniquely attenuate NMDAR-mediated currents, limit calcium influx, and suppress dendritic spine formation, high levels during early childhood may be important for regulating neuroprotection and activity-dependent sculpting of synapses. We also examined whether subunit changes underlie reduced NMDAR activity in schizophrenia. Our results reveal normal NR1 and NR3A protein levels in DLPFC from schizophrenic patients, indicating that NMDAR hypofunction is unlikely to be maintained by gross changes in NR3A-containing NMDARs or overall NMDAR numbers. These data provide insights into NMDAR functions in the developing CNS and will contribute to designing pharmacotherapies for neurological disorders.
Collapse
Affiliation(s)
| | - Adam C. Roberts
- Neuroscience Center
- Department of Cell and Molecular Physiology
| | | | | | - Robert M. Hamer
- Department of Psychiatry
- Schizophrenia Research Center
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27705, USA
| | - John H. Gilmore
- Curriculum in Neurobiology
- Department of Psychiatry
- Schizophrenia Research Center
| | - L. Fredrik Jarskog
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Benjamin D. Philpot
- Curriculum in Neurobiology
- Neuroscience Center
- Department of Cell and Molecular Physiology
| |
Collapse
|
44
|
Abstract
Brain-derived neurotrophic factor (BDNF) has emerged as a regulator of development, plasticity and, recently, addiction. Decreased neurotrophic activity may be involved in ethanol-induced neurodegeneration in the adult brain and in the etiology of alcohol-related neurodevelopmental disorders. This can occur through decreased expression of BDNF or through inability of the receptor to transduce signals in the presence of ethanol. In contrast, recent studies implicate region-specific up-regulation of BDNF and associated signaling pathways in anxiety, addiction and homeostasis after ethanol exposure. Anxiety and depression are precipitating factors for substance abuse and these disorders also involve region-specific changes in BDNF in both pathogenesis and response to pharmacotherapy. Polymorphisms in the genes coding for BDNF and its receptor TrkB are linked to affective, substance abuse and appetitive disorders and therefore may play a role in the development of alcoholism. This review summarizes historical and pre-clinical data on BDNF and TrkB as it relates to ethanol toxicity and addiction. Many unresolved questions about region-specific changes in BDNF expression and the precise role of BDNF in neuropsychiatric disorders and addiction remain to be elucidated. Resolution of these questions will require significant integration of the literature on addiction and comorbid psychiatric disorders that contribute to the development of alcoholism.
Collapse
Affiliation(s)
- Margaret I Davis
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Nagy J. Alcohol related changes in regulation of NMDA receptor functions. Curr Neuropharmacol 2008; 6:39-54. [PMID: 19305787 PMCID: PMC2645546 DOI: 10.2174/157015908783769662] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 07/19/2007] [Accepted: 10/20/2007] [Indexed: 12/25/2022] Open
Abstract
Long-term alcohol exposure may lead to development of alcohol dependence in consequence of altered neurotransmitter functions. Accumulating evidence suggests that the N-methyl-D-aspartate (NMDA) type of glutamate receptors is a particularly important site of ethanol's action. Several studies showed that ethanol potently inhibits NMDA receptors (NMDARs) and prolonged ethanol exposition leads to a compensatory "up-regulation" of NMDAR mediated functions. Therefore, alterations in NMDAR function are supposed to contribute to the development of ethanol tolerance, dependence as well as to the acute and late signs of ethanol withdrawal.A number of publications report alterations in the expression and phosphorylation states of NMDAR subunits, in their interaction with scaffolding proteins or other receptors in consequence of chronic ethanol treatment. Our knowledge on the regulatory processes, which modulate NMDAR functions including factors altering transcription, protein expression and post-translational modifications of NMDAR subunits, as well as those influencing their interactions with different regulatory proteins or other downstream signaling elements are incessantly increasing. The aim of this review is to summarize the complex chain of events supposedly playing a role in the up-regulation of NMDAR functions in consequence of chronic ethanol exposure.
Collapse
Affiliation(s)
- József Nagy
- Gedeon Richter Plc., Pharmacological and Drug Safety Research, Laboratory for Molecular Cell Biology, Budapest 10. P.O. Box 27, H-1475 Hungary.
| |
Collapse
|
46
|
Knapp DJ, Overstreet DH, Angel RA, Navarro M, Breese GR. The amygdala regulates the antianxiety sensitization effect of flumazenil during repeated chronic ethanol or repeated stress. Alcohol Clin Exp Res 2007; 31:1872-82. [PMID: 17908266 PMCID: PMC2864652 DOI: 10.1111/j.1530-0277.2007.00514.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The benzodiazepine receptor antagonist flumazenil reduces anxiety-like behavior and sensitization of anxiety-like behavior in various models of ethanol withdrawal in rodents. The mechanism and brain region(s) that account for this action of flumazenil remain unknown. This investigation explored the potential role of several brain regions (amygdala, raphe, inferior colliculus, nucleus accumbens, and paraventricular hypothalamus) for these actions of flumazenil. METHODS Rats were surgically implanted with guide cannulae directed over the brain region of interest and then treated with an ethanol diet for three 7-day dietary cycles (5 days on ethanol diet followed by 2 days on control diet). At approximately 4 hours, flumazenil was administered intracranially into each of the first 2 withdrawals. Examinations of anxiety-like behavior followed 1 week later during a third withdrawal. In other animals, restraint stress sessions or intra-amygdala DMCM (methyl-6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate) injections, preceded by intraperitoneal flumazenil injections, were substituted for the first 2 ethanol treatment cycles to assess the potential anxiety-sensitizing action of stress or a benzodiazepine receptor inverse agonist, respectively. RESULTS Flumazenil treatment of the amygdala during the first 2 withdrawals blocked the development of sensitized anxiety seen during a third withdrawal. Similar actions of flumazenil were found when stress sessions substituted for the first 2 cycles of ethanol exposure and withdrawal. Amygdala treatment with DMCM magnified the anxiety response to the single subthreshold chronic ethanol treatment, and prophylactic flumazenil blocked this effect. CONCLUSIONS Intra-amygdala flumazenil inhibits the development of anxiety sensitized by repeated ethanol withdrawal, stress/ethanol withdrawal, or DMCM/ethanol withdrawal. These actions suggest that site-specific and persistent effects of flumazenil on gamma-aminobutyric acid-modulatory processes in this brain region are relevant to sensitized behavioral effects seen in alcoholism.
Collapse
Affiliation(s)
- Darin J Knapp
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7158, USA.
| | | | | | | | | |
Collapse
|
47
|
Van Kolen K, Pullan S, Neefs JM, Dautzenberg FM. Nociceptive and behavioural sensitisation by protein kinase Cepsilon signalling in the CNS. J Neurochem 2007; 104:1-13. [PMID: 17971128 DOI: 10.1111/j.1471-4159.2007.04986.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the apparent homology in the protein kinase C (PKC) family, it has become clear that slight structural differences are sufficient to have unique signalling properties for each individual isoform. For PKCepsilon in depth investigation of these aspects revealed unique actions in the CNS and lead to development of specific modulators with clinical perspective. In this review, we describe to which extent PKCepsilon is distinct from other isoforms on the level of tissue expression and protein structure. As this kinase is highly expressed in the brain, we outline three main aspects of PKCepsilon signalling in the CNS. First, its ability to alter the permeability of N-type Ca2+ channels in dorsal root ganglia has been shown to enhance nociception. Secondly, PKCepsilon increases anxiety by diminishing GABA(A)R-induced inhibitory post-synaptic currents in the prefrontal cortex. Another important aspect of the latter inhibition is the reduced sensitivity of GABA(A) receptors to ethanol, a mechanism potentially contributing to abuse. A third signalling cascade improves cognitive functions by facilitating cholinergic signalling in the hippocampus. Collectively, these findings point to a physical and behavioural sensitising role for this kinase.
Collapse
Affiliation(s)
- Kristof Van Kolen
- CNS Research, Johnson & Johnson Pharmaceutical Research and Development, Beerse, Belgium.
| | | | | | | |
Collapse
|
48
|
Rudoy CA, Van Bockstaele EJ. Betaxolol, a selective beta(1)-adrenergic receptor antagonist, diminishes anxiety-like behavior during early withdrawal from chronic cocaine administration in rats. Prog Neuropsychopharmacol Biol Psychiatry 2007; 31:1119-29. [PMID: 17513029 PMCID: PMC4287233 DOI: 10.1016/j.pnpbp.2007.04.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 03/16/2007] [Accepted: 04/09/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Anxiety has been indicated as one of the main symptoms of the cocaine withdrawal syndrome in human addicts and severe anxiety during withdrawal may potentially contribute to relapse. As alterations in noradrenergic transmission in limbic areas underlie withdrawal symptomatology for many drugs of abuse, the present study sought to determine the effect of cocaine withdrawal on beta-adrenergic receptor (beta(1) and beta(2)) expression in the amygdala. METHODS Male Sprague Dawley rats were administered intraperitoneal (i.p.) injections of cocaine (20 mg/kg) once daily for 14 days. Two days following the last cocaine injection, amygdala brain regions were micro-dissected and processed for Western blot analysis. Results showed that beta(1)-adrenergic receptor, but not beta(2)-adrenergic receptor expression was significantly increased in amygdala extracts of cocaine-withdrawn animals as compared to controls. This finding motivated further studies aimed at determining whether treatment with betaxolol, a highly selective beta(1)-adrenergic receptor antagonist, could ameliorate cocaine withdrawal-induced anxiety. In these studies, betaxolol (5 mg/kg via i.p. injection) was administered at 24 and then 44 h following the final chronic cocaine administration. Anxiety-like behavior was evaluated using the elevated plus maze test approximately 2 h following the last betaxolol injection. Following behavioral testing, betaxolol effects on beta(1)-adrenergic receptor protein expression were examined by Western blotting in amygdala extracts from rats undergoing cocaine withdrawal. RESULTS Animals treated with betaxolol during cocaine withdrawal exhibited a significant attenuation of anxiety-like behavior characterized by increased time spent in the open arms and increased entries into the open arms compared to animals treated with only saline during cocaine withdrawal. In contrast, betaxolol did not produce anxiolytic-like effects in control animals treated chronically with saline. Furthermore, treatment with betaxolol during early cocaine withdrawal significantly decreased beta(1)-adrenergic receptor protein expression in the amygdala to levels comparable to those of control animals. CONCLUSIONS The present findings suggest that the anxiolytic-like effect of betaxolol on cocaine-induced anxiety may be related to its effect on amygdalar beta(1)-adrenergic receptors that are up-regulated during early phases of drug withdrawal. These data support the efficacy of betaxolol as a potential effective pharmacotherapy in treating cocaine withdrawal-induced anxiety during early phases of abstinence.
Collapse
MESH Headings
- Adrenergic beta-Antagonists/therapeutic use
- Amygdala/drug effects
- Amygdala/metabolism
- Animals
- Anxiety/drug therapy
- Anxiety/etiology
- Anxiety/psychology
- Behavior, Animal/drug effects
- Betaxolol/therapeutic use
- Blotting, Western
- Chronic Disease
- Cocaine-Related Disorders/psychology
- Dose-Response Relationship, Drug
- Male
- Motor Activity/drug effects
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, beta-1/biosynthesis
- Receptors, Adrenergic, beta-1/drug effects
- Receptors, Adrenergic, beta-2/drug effects
- Substance Withdrawal Syndrome/drug therapy
- Substance Withdrawal Syndrome/psychology
Collapse
Affiliation(s)
- C A Rudoy
- Department of Neurosurgery, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | |
Collapse
|
49
|
Oliva JM, Manzanares J. Gene transcription alterations associated with decrease of ethanol intake induced by naltrexone in the brain of Wistar rats. Neuropsychopharmacology 2007; 32:1358-69. [PMID: 17063152 DOI: 10.1038/sj.npp.1301237] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Preclinical and clinical studies suggest that the administration of the opioid antagonist naltrexone decreases the intake of ethanol. However, the neuroplastic adaptations in the brain associated to reduction of ethanol consumption remains to be elucidated. The aim of the study was to identify gene transcription alterations underlying the attenuation of voluntary ethanol intake by administration of naltrexone in rats. Increasing doses of naltrexone (0.7 mg/kg, 4 days and 1.4 mg/kg/day, 4 days) to rats with acquired high preferring ethanol consumption (>3.5 g of ethanol/kg/day) decreased voluntary ethanol intake (50%). Voluntary ethanol consumption altered mu-opioid receptor function in the cingulate cortex, caudate-putamen (CPu), nucleus accumbens core (Acb C) and shell (Acb S), the expression of tyrosine hydroxylase (TH) in the ventral tegmental area and substantia nigra, proenkephalin (PENK) in the piriform cortex, olfactory tubercle, CPu, Acb C and Acb S, ventromedial nucleus (VMN) and paraventricular nucleus (PVN) of the hypothalamus, corticotropin releasing factor (CRF) in PVN, cannabinoid CB(1) receptor (CB1-R) in the CPu, hippocampus and VMN, and serotonin transporter (5-HTT) in the dorsal and median raphe nuclei. The reduction of ethanol intake induced by naltrexone was associated with a blockade or significant reduction of the changes produced by ethanol in the expression of these genes in key regions related to drug dependence. These results point to a role for the mu-opioid receptor, TH, PENK, CRF, CB1-R, and 5-HTT genes in specific brain regions in the modulation of neuroadaptative mechanisms associated to the decrease of ethanol intake induced by naltrexone.
Collapse
Affiliation(s)
- José M Oliva
- Institute of Physiological Chemistry and Pathobiochemistry, School of Medicine, Johannes-Gutemberg University of Mainz, Mainz, Germany
| | | |
Collapse
|