1
|
Uusi-Oukari M, Korpi ER. GABAergic mechanisms in alcohol dependence. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:75-123. [PMID: 38555121 DOI: 10.1016/bs.irn.2024.03.002] [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: 04/02/2024]
Abstract
The target of alcohol's effect on the central nervous system has been sought for more than 50 years in the brain's GABA system. The behavioral and emotional effects of alcohol in humans and rodents are very similar to those of barbiturates and benzodiazepines, and GABAA receptors have been shown to be one of the sites of alcohol action. The mechanisms of GABAergic inhibition have been a hotspot of research but have turned out to be complex and controversial. Genetics support the involvement of some GABAA receptor subunits in the development of alcohol dependence and in alcohol use disorders (AUD). Since the effect of alcohol on the GABAA system resembles that of a GABAergic positive modulator, it may be possible to develop GABAergic drug treatments that could substitute for alcohol. The adaptation mechanisms of the GABA system and the plasticity of the brain are a big challenge for drug development: the drugs that act on GABAA receptors developed so far also may cause adaptation and development of additional addiction. Human polymorphisms should be studied further to get insight about how they affect receptor function, expression or other factors to make reasonable predictions/hypotheses about what non-addictive interventions would help in alcohol dependence and AUD.
Collapse
Affiliation(s)
- Mikko Uusi-Oukari
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Qin J, Wei T, Chen H, Lin X, Qin D, Wei F, Liu P, Ye W, Su J. Salicylate Induced GABAAR Internalization by Dopamine D1-Like Receptors Involving Protein Kinase C (PKC) in Spiral Ganglion Neurons. Med Sci Monit 2021; 27:e933278. [PMID: 34657931 PMCID: PMC8532520 DOI: 10.12659/msm.933278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Sodium salicylate (SS) induces excitotoxicity of spiral ganglion neurons (SGNs) by inhibiting the response of γ-aminobutyric acid type A receptors (GABAARs). Our previous studies have shown that SS can increase the internalization of GABAARs on SGNs, which involves dopamine D1-like receptors (D1Rs) and related signaling pathways. In this study, we aimed to explore the role of D1Rs and their downstream molecule protein kinase C (PKC) in the process of SS inhibiting GABAARs. MATERIAL AND METHODS The expression of D1Rs and GABARγ2 on rat cochlear SGNs cultured in vitro was tested by immunofluorescence. Then, the SGNs were exposed to SS, D1R agonist (SKF38393), D1R antagonist (SCH23390), clathrin/dynamin-mediated endocytosis inhibitor (dynasore), and PKC inhibitor (Bisindolylmaleimide I). Western blotting and whole-cell patch clamp technique were used to assess the changes of surface and total protein of GABARγ2 and GABA-activated currents. RESULTS Immunofluorescence showed that D1 receptors (DRD1) were expressed on SGNs. Data from western blotting showed that SS promoted the internalization of cell surface GABAARs, and activating D1Rs had the same result. Inhibiting D1Rs and PKC decreased the internalization of GABAARs. Meanwhile, the phosphorylation level of GABAARγ2 S327 affected by PKC was positively correlated with the degree of internalization of GABAARs. Moreover, whole-cell patch clamp recording showed that inhibition of D1Rs or co-inhibition of D1Rs and PKC attenuated the inhibitory effect of SS on GABA-activated currents. CONCLUSIONS D1Rs mediate the GABAAR internalization induced by SS via a PKC-dependent manner and participate in the excitotoxic process of SGNs.
Collapse
Affiliation(s)
- Jiangyuan Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Tingjia Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Huiying Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xiaoyu Lin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Fangyu Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Wenhua Ye
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiping Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
3
|
Barker JS, Hines RM. Regulation of GABA A Receptor Subunit Expression in Substance Use Disorders. Int J Mol Sci 2020; 21:ijms21124445. [PMID: 32580510 PMCID: PMC7352578 DOI: 10.3390/ijms21124445] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 01/02/2023] Open
Abstract
The modulation of neuronal cell firing is mediated by the release of the neurotransmitter GABA (γ-aminobuytric acid), which binds to two major families of receptors. The ionotropic GABAA receptors (GABAARs) are composed of five distinct subunits that vary in expression by brain region and cell type. The action of GABA on GABAARs is modulated by a variety of clinically and pharmacologically important drugs such as benzodiazepines and alcohol. Exposure to and abuse of these substances disrupts homeostasis and induces plasticity in GABAergic neurotransmission, often via the regulation of receptor expression. Here, we review the regulation of GABAAR subunit expression in adaptive and pathological plasticity, with a focus on substance use. We examine the factors influencing the expression of GABAAR subunit genes including the regulation of the 5′ and 3′ untranslated regions, variations in DNA methylation, immediate early genes and transcription factors that regulate subunit expression, translational and post-translational modifications, and other forms of receptor regulation beyond expression. Advancing our understanding of the factors regulating GABAAR subunit expression during adaptive plasticity, as well as during substance use and withdrawal will provide insight into the role of GABAergic signaling in substance use disorders, and contribute to the development of novel targeted therapies.
Collapse
|
4
|
Hughes BA, Bohnsack JP, O'Buckley TK, Herman MA, Morrow AL. Chronic Ethanol Exposure and Withdrawal Impair Synaptic GABA A Receptor-Mediated Neurotransmission in Deep-Layer Prefrontal Cortex. Alcohol Clin Exp Res 2019; 43:822-832. [PMID: 30860602 DOI: 10.1111/acer.14015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/05/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND The prefrontal cortex (PFC) acts as an integrative hub for the processing of cortical and subcortical input into meaningful efferent signaling, permitting complex associative behaviors. PFC dysfunction is consistently observed with ethanol (EtOH) dependence and is a core component of the pathology of alcohol use disorders in current models of addiction. While intracortical gamma-aminobutryric acid (GABA)ergic neurotransmission is understood to be essential for maintaining coordinated network activity within the cortex, relatively little is known regarding functional GABAergic adaptations in PFC during EtOH dependence. METHODS In the present study, male and female (> postnatal day 60) Sprague-Dawley rats were administered EtOH (5.0 g/kg; intragastric gavage) for 14 to 15 consecutive days. Twenty-four hours after the final administration, animals were sacrificed and brains extracted for electrophysiological recordings of isolated GABAA receptor-mediated currents or analysis of GABAA receptor subunit protein expression in deep-layer PFC neurons. RESULTS Chronic EtOH exposure significantly attenuated activity-dependent spontaneous GABAA receptor-mediated inhibitory postsynaptic current (IPSC) frequency with no effect on amplitude. Furthermore, analysis of IPSC decay kinetics revealed a significant enhancement of IPSC decay time that was associated with decrements in expression of the α1 GABAA receptor subunit, indicative of further impaired phasic inhibition. These phenomena occurred irrespective of neuron projection destination and sex. Based on previous observations by our laboratory of an epigenetic mechanism for EtOH-induced changes in cortical GABAA receptor subunit expression, the histone deacetylase inhibitor Trichostatin A was administered to water- and EtOH-exposed animals, and prevented EtOH-induced changes in spontaneous IPSC frequency, IPSC decay kinetics, and GABAA receptor subunit expression. CONCLUSIONS Taken together, these results demonstrate that chronic EtOH exposure impairs synaptic inhibitory neurotransmission in deep-layer pyramidal neurons of the medial PFC in both male and female rats. These maladaptations occur in neurons projecting to numerous regions implicated in the sequelae of EtOH dependence, offering a mechanistic link between the manifestation of PFC dysfunction and negative affective states observed with extended consumption.
Collapse
Affiliation(s)
- Benjamin A Hughes
- Department of Psychiatry , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Bowles Center for Alcohol Studies , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John Peyton Bohnsack
- Department of Pharmacology , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Bowles Center for Alcohol Studies , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Todd K O'Buckley
- Bowles Center for Alcohol Studies , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A Herman
- Department of Pharmacology , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Bowles Center for Alcohol Studies , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - A Leslie Morrow
- Department of Psychiatry , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Bowles Center for Alcohol Studies , School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
5
|
Fontana BD, Stefanello FV, Mezzomo NJ, Müller TE, Quadros VA, Parker MO, Rico EP, Rosemberg DB. Taurine modulates acute ethanol-induced social behavioral deficits and fear responses in adult zebrafish. J Psychiatr Res 2018; 104:176-182. [PMID: 30096615 DOI: 10.1016/j.jpsychires.2018.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/25/2018] [Accepted: 08/02/2018] [Indexed: 01/08/2023]
Abstract
Ethanol (EtOH) is a central nervous system (CNS) depressant drug that modifies various behavioral domains (i.e., sociability, aggressiveness, and memory) by promoting disinhibition of punished operant behavior and neurochemical changes. Taurine (TAU) is a β-amino sulfonic acid with pleiotropic roles in the brain. Although exogenous TAU is found in energy drinks and often mixed with alcohol in beverages, the putative risks of mixing TAU and EtOH are poorly explored. Here, we investigated whether TAU modulates social and fear responses by assessing shoaling behavior, preference for conspecifics, and antipredatory behavior of adult zebrafish acutely exposed to EtOH. Zebrafish shoals (4 fish per shoal) were exposed to water (control), TAU (42, 150, and 400 mg/L), 0.25% (v/v) EtOH alone or in association with TAU for 1 h, and their behaviors were analyzed at different time intervals (0-5 min, 30-35 min, and 55-60 min). The effects of TAU and EtOH were further tested in a social preference test and during exposure to a predator. Both EtOH and TAU co-treated fish showed a higher shoal dispersion, while TAU 400/EtOH group shoal area had a similar profile when compared to control. However, in the social preference test, TAU 400/EtOH impaired the seeking for conspecifics. Regarding fear-like behaviors, TAU-cotreated fish showed a prominent reduction in risk assessments when compared to EtOH alone. Overall, we demonstrate that TAU modulates EtOH-induced changes in different behavioral domains, suggesting a complex relationship between social and fear-like responses.
Collapse
Affiliation(s)
- Barbara D Fontana
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil.
| | - Flavia V Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Nathana J Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Talise E Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Vanessa A Quadros
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA
| | - Eduardo P Rico
- Graduate Program in Health Sciences, Laboratory of Neural Signaling and Psychopharmacology, Academic Unit of Health Sciences, University of Southern Santa Catarina (UNESC), 1105, Universitária Avenue, Bloco S, Sala 6, Criciúma, SC, 88806-000, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA.
| |
Collapse
|
6
|
Gravielle MC. Regulation of GABAA receptors by prolonged exposure to endogenous and exogenous ligands. Neurochem Int 2018; 118:96-104. [DOI: 10.1016/j.neuint.2018.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 02/08/2023]
|
7
|
Histone deacetylases mediate GABA A receptor expression, physiology, and behavioral maladaptations in rat models of alcohol dependence. Neuropsychopharmacology 2018; 43. [PMID: 29520058 PMCID: PMC5983537 DOI: 10.1038/s41386-018-0034-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Alcohol use disorders are chronic debilitating diseases characterized by severe withdrawal symptoms that contribute to morbidity and relapse. GABAA receptor (GABAAR) adaptations have long been implicated in the chronic effects of alcohol and contribute to many withdrawal symptoms associated with alcohol dependence. In rodents, GABAAR hypofunction results from decreases in Gabra1 expression, although the underlying mechanism controlling Gabra1 expression after chronic ethanol exposure is still unknown. We found that chronic ethanol exposure using either ethanol gavage or two-bottle choice voluntary access paradigms decreased Gabra1 expression and increased Hdac2 and Hdac3 expression. Administration of the HDAC inhibitor trichostatin A (TSA) after chronic ethanol exposure prevents the decrease in Gabra1 expression and function as well as the increase in Hdac2 and Hdac3 expression in both the cortex and the medial prefrontal cortex (mPFC). Chronic ethanol exposure and withdrawal, but not acute ethanol exposure or acute withdrawal, cause a selective upregulation of HDAC2 and HDAC3 associated with the Gabra1 promoter that accompanies a decrease in H3 acetylation of the Gabra1 promoter and the reduction in GABAAR α1 subunit expression. TSA administration prevented each of these molecular events as well as behavioral manifestations of ethanol dependence, including tolerance to zolpidem-induced loss of righting reflex, reduced open-arm time in the elevated plus maze, reduced center-time and locomotor activity in the open-field assay, and TSA reduced voluntary ethanol consumption. The results show how chronic ethanol exposure regulates the highly prominent GABAAR α1 subunit by an epigenetic mechanism that represents a potential treatment modality for alcohol dependence.
Collapse
|
8
|
Chen J, He Y, Wu Y, Zhou H, Su LD, Li WN, Olsen RW, Liang J, Zhou YD, Shen Y. Single Ethanol Withdrawal Regulates Extrasynaptic δ-GABA A Receptors Via PKCδ Activation. Front Mol Neurosci 2018; 11:141. [PMID: 29755316 PMCID: PMC5932167 DOI: 10.3389/fnmol.2018.00141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/09/2018] [Indexed: 11/23/2022] Open
Abstract
Alcohol (ethanol, EtOH) is one of the most widely abused drugs with profound effects on brain function and behavior. GABAA receptors (GABAARs) are one of the major targets for EtOH in the brain. Temporary plastic changes in GABAARs after withdrawal from a single EtOH exposure occur both in vivo and in vitro, which may be the basis for chronic EtOH addiction, tolerance and withdrawal symptoms. Extrasynaptic δ-GABAAR endocytosis is implicated in EtOH-induced GABAAR plasticity, but the mechanisms by which the relative abundance and localization of specific GABAARs are altered by EtOH exposure and withdrawal remain unclear. In this study, we investigated the mechanisms underlying rapid regulation of extrasynaptic δ-GABAAR by a single EtOH withdrawal in cultured rat hippocampal neurons. Thirty-minutes EtOH (60 mM) exposure increased extrasynaptic tonic current (Itonic) amplitude without affecting synaptic GABAAR function in neurons. In contrast, at 30 min after withdrawal, Itonic amplitude and responsiveness to acute EtOH were both reduced. Similar results occurred in neurons with okadaic acid (OA) or phorbol 12,13-dibutyrate (PDBu) exposure. Protein kinase C (PKC) inhibition prevented the reduction of Itonic amplitude and the tolerance to acute EtOH, as well as the reduction of GABAAR-δ subunit abundance induced by a single EtOH withdrawal. Moreover, EtOH withdrawal selectively increased PKCδ level, whereas PKCδ inhibition specifically rescued the EtOH-induced alterations in GABAAR-δ subunit level and δ-GABAAR function. Together, we provided strong evidence for the important roles of PKCδ in the rapid regulation of extrasynaptic δ-GABAAR induced by a single EtOH withdrawal.
Collapse
Affiliation(s)
- Juan Chen
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang He
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zhou
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Nan Li
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Richard W Olsen
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jing Liang
- Titus Family Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Yu-Dong Zhou
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Shen
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Lorenz-Guertin JM, Jacob TC. GABA type a receptor trafficking and the architecture of synaptic inhibition. Dev Neurobiol 2018; 78:238-270. [PMID: 28901728 PMCID: PMC6589839 DOI: 10.1002/dneu.22536] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
Ubiquitous expression of GABA type A receptors (GABAA R) in the central nervous system establishes their central role in coordinating most aspects of neural function and development. Dysregulation of GABAergic neurotransmission manifests in a number of human health disorders and conditions that in certain cases can be alleviated by drugs targeting these receptors. Precise changes in the quantity or activity of GABAA Rs localized at the cell surface and at GABAergic postsynaptic sites directly impact the strength of inhibition. The molecular mechanisms constituting receptor trafficking to and from these compartments therefore dictate the efficacy of GABAA R function. Here we review the current understanding of how GABAA Rs traffic through biogenesis, plasma membrane transport, and degradation. Emphasis is placed on discussing novel GABAergic synaptic proteins, receptor and scaffolding post-translational modifications, activity-dependent changes in GABAA R confinement, and neuropeptide and neurosteroid mediated changes. We further highlight modern techniques currently advancing the knowledge of GABAA R trafficking and clinically relevant neurodevelopmental diseases connected to GABAergic dysfunction. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 238-270, 2018.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| |
Collapse
|
10
|
Popoola DO, Cameron NM. Maternal care-related differences in males and females rats' sensitivity to ethanol and the associations between the GABAergic system and steroids in males. Dev Psychobiol 2018; 60:380-394. [PMID: 29442358 DOI: 10.1002/dev.21607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
Abstract
This study investigated the effect of maternal care on adolescent ethanol consumption, sensitivity to ethanol-induced hypnosis, as well as gonadal hormones and γ-aminobutyric acid type-A (GABAA ) systems. Long Evans rat dams were categorized by maternal licking/grooming (LG) frequency into High- and Low-LG mothers. Both female and male offspring from Low-LG rats demonstrated a greater sensitivity to ethanol-induced hypnosis in the loss-of-righting-reflex test at ethanol doses of 3.0 and 3.5 g/kg during late-adolescence (postnatal Day 50) but not at mid-adolescence (postnatal Day 42). However, we found no effect of maternal care on consumption of a 5% ethanol solution in a two-bottle choice test. We further investigated the association between the observed variations in sensitivity to ethanol-induced hypnosis and baseline hormonal levels in males. In male offspring from Low-LG mothers compared to High-LG mothers, baseline plasma corticosterone and progesterone levels were higher. GABAA α1 and δ subunit expressions were also higher in the cerebral cortex of Low-LG males but lower in the cerebellar synaptosomal fraction. Early environmental influences on adolescent sensitivity to ethanol-induced hypnosis, consumption, and preference may be mediated by gonadal hormones and possibly through GABAergic functions.
Collapse
Affiliation(s)
- Daniel O Popoola
- Department of Psychology, Center for Developmental and Behavioral Neuroscience, Binghamton University, Binghamton, New York.,Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, New York.,Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University of Buffalo, Buffalo, New York
| | - Nicole M Cameron
- Department of Psychology, Center for Developmental and Behavioral Neuroscience, Binghamton University, Binghamton, New York.,Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, New York
| |
Collapse
|
11
|
Lorenz-Guertin JM, Wilcox MR, Zhang M, Larsen MB, Pilli J, Schmidt BF, Bruchez MP, Johnson JW, Waggoner AS, Watkins SC, Jacob TC. A versatile optical tool for studying synaptic GABA A receptor trafficking. J Cell Sci 2017; 130:3933-3945. [PMID: 29025969 DOI: 10.1242/jcs.205286] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/26/2017] [Indexed: 12/26/2022] Open
Abstract
Live-cell imaging methods can provide critical real-time receptor trafficking measurements. Here, we describe an optical tool to study synaptic γ-aminobutyric acid (GABA) type A receptor (GABAAR) dynamics through adaptable fluorescent-tracking capabilities. A fluorogen-activating peptide (FAP) was genetically inserted into a GABAAR γ2 subunit tagged with pH-sensitive green fluorescent protein (γ2pHFAP). The FAP selectively binds and activates Malachite Green (MG) dyes that are otherwise non-fluorescent in solution. γ2pHFAP GABAARs are expressed at the cell surface in transfected cortical neurons, form synaptic clusters and do not perturb neuronal development. Electrophysiological studies show γ2pHFAP GABAARs respond to GABA and exhibit positive modulation upon stimulation with the benzodiazepine diazepam. Imaging studies using γ2pHFAP-transfected neurons and MG dyes show time-dependent receptor accumulation into intracellular vesicles, revealing constitutive endosomal and lysosomal trafficking. Simultaneous analysis of synaptic, surface and lysosomal receptors using the γ2pHFAP-MG dye approach reveals enhanced GABAAR turnover following a bicucculine-induced seizure paradigm, a finding not detected by standard surface receptor measurements. To our knowledge, this is the first application of the FAP-MG dye system in neurons, demonstrating the versatility to study nearly all phases of GABAAR trafficking.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Madeleine R Wilcox
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ming Zhang
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Mads B Larsen
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jyotsna Pilli
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Brigitte F Schmidt
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Marcel P Bruchez
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jon W Johnson
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Alan S Waggoner
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Simon C Watkins
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
12
|
Bohnsack JP, Patel VK, Morrow AL. Ethanol Exposure Regulates Gabra1 Expression via Histone Deacetylation at the Promoter in Cultured Cortical Neurons. J Pharmacol Exp Ther 2017; 363:1-11. [PMID: 28798030 PMCID: PMC5596976 DOI: 10.1124/jpet.117.242446] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/08/2017] [Indexed: 12/23/2022] Open
Abstract
γ-Aminobutyric acid A receptors (GABAA-Rs) mediate the majority of inhibitory neurotransmission in the adult brain. The α1-containing GABAA-Rs are the most prominent subtype in the adult brain and are important in both homeostatic function and several disease pathologies including alcohol dependence, epilepsy, and stress. Ethanol exposure causes a decrease of α1 transcription and peptide expression both in vivo and in vitro, but the mechanism that controls the transcriptional regulation is unknown. Because ethanol is known to activate epigenetic regulation of gene expression, we tested the hypothesis that ethanol regulates α1 expression through histone modifications in cerebral cortical cultured neurons. We found that class I histone deacetylases (HDACs) regulate ethanol-induced changes in α1 gene and protein expression as pharmacologic inhibition or knockdown of HDAC1-3 prevents the effects of ethanol exposure. Targeted histone acetylation associated with the Gabra1 promoter using CRISPR (clustered regularly interspaced palindromic repeat) dCas9-P300 (a nuclease-null Cas9 fused with a histone acetyltransferase) increases histone acetylation and prevents the decrease of Gabra1 expression. In contrast, there was no effect of a mutant histone acetyltransferase or generic transcriptional activator or targeting P300 to a distant exon. Conversely, using a dCas9-KRAB construct that increases repressive methylation (H3K9me3) does not interfere with ethanol-induced histone deacetylation. Overall our results indicate that ethanol deacetylates histones associated with the Gabra1 promoter through class I HDACs and that pharmacologic, genetic, or epigenetic intervention prevents decreases in α1 expression in cultured cortical neurons.
Collapse
Affiliation(s)
- John Peyton Bohnsack
- Department of Pharmacology (J.P.B., A.L.M.), Department of Psychiatry (A.L.M.), and Bowles Center for Alcohol Studies (J.P.B., V.K.P., A.L.M.), University of North Carolina, Chapel Hill, North Carolina
| | - Vraj K Patel
- Department of Pharmacology (J.P.B., A.L.M.), Department of Psychiatry (A.L.M.), and Bowles Center for Alcohol Studies (J.P.B., V.K.P., A.L.M.), University of North Carolina, Chapel Hill, North Carolina
| | - A Leslie Morrow
- Department of Pharmacology (J.P.B., A.L.M.), Department of Psychiatry (A.L.M.), and Bowles Center for Alcohol Studies (J.P.B., V.K.P., A.L.M.), University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
13
|
Rebai O, Belkhir M, Sanchez-Gomez MV, Matute C, Fattouch S, Amri M. Differential Molecular Targets for Neuroprotective Effect of Chlorogenic Acid and its Related Compounds Against Glutamate Induced Excitotoxicity and Oxidative Stress in Rat Cortical Neurons. Neurochem Res 2017; 42:3559-3572. [PMID: 28948515 DOI: 10.1007/s11064-017-2403-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/19/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
Abstract
The present study has been designed to explore the molecular mechanism and signaling pathway targets of chlorogenic acid (CGA) and its main hydrolysates, caffeic (CA) and quinic acid in the protective effect against glutamate-excitotoxicity. For this purpose 8-DIV cortical neurons in primary culture were exposed to 50 μM L-glutamic acid plus 10 µM glycine, with or without 10-100 μM tested compounds. Chlorogenic acid and caffeic acid via their antioxidant properties inhibited cell death induced by glutamate in dose depended manner. However, quinic acid slightly protects neurons at a higher dose. DCF, JC-1 and Ca2+sensitive fluorescent dye fura-2, were used to measure intracellular ROS accumulation, mitochondrial membrane potential integration and intracellular calcium concentration [Ca2+] i . Results indicate that similarly, CGA acts as a protective agent against glutamate-induced cortical neurons injury by suppressing the accumulation of endogenous ROS and restore the mitochondrial membrane potential, activate the enzymatic antioxidant system by the increase levels of SOD activity and modulate the rise of intracellular calcium levels by increasing the rise of intracellular concentrations of Ca2+caused by glutamate overstimulation. PKC signaling cascade appear to be engaged in this protective mechanism. Interseling, CGA and CA also exhibit antiapoptotic properties against glutamate-induced cleaved activation of pro-caspases; caspase 1,8 and 9 and calpain (PD 150606,Calpeptin and MDL 28170).These data suggest that neuroprotective activity of CGA ester may occurs throught its hydrolysate,the caffeic acid and its interaction with intracellular molecules suggesting that CGA exert its neuroprotection via its caffeoly acid group that might potentially be used as a therapeutic agent in neurodegeneratives disorders associated with glutamate excitotoxicity.
Collapse
Affiliation(s)
- Olfa Rebai
- Research Unit of Functional Neurophysiology and Pathology, 00/UR/08-01, Department of Biological Sciences, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia.
| | - Manel Belkhir
- Research Unit of Functional Neurophysiology and Pathology, 00/UR/08-01, Department of Biological Sciences, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - María Victoria Sanchez-Gomez
- Departamento de Neurociencias, Facultad de Medicina y Odontologia, Universidad Del Paıs Vasco, Leioa, Vizcaya, Spain
| | - Carlos Matute
- Departamento de Neurociencias, Facultad de Medicina y Odontologia, Universidad Del Paıs Vasco, Leioa, Vizcaya, Spain
| | - Sami Fattouch
- Laboratoire de Biochimie Alimentaire, INSAT, University of Carthage, Tunis, Tunisia
| | - Mohamed Amri
- Research Unit of Functional Neurophysiology and Pathology, 00/UR/08-01, Department of Biological Sciences, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| |
Collapse
|
14
|
van der Vaart AD, Wolstenholme JT, Smith ML, Harris GM, Lopez MF, Wolen AR, Becker HC, Williams RW, Miles MF. The allostatic impact of chronic ethanol on gene expression: A genetic analysis of chronic intermittent ethanol treatment in the BXD cohort. Alcohol 2017; 58:93-106. [PMID: 27838001 DOI: 10.1016/j.alcohol.2016.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 11/25/2022]
Abstract
The transition from acute to chronic ethanol exposure leads to lasting behavioral and physiological changes such as increased consumption, dependence, and withdrawal. Changes in brain gene expression are hypothesized to underlie these adaptive responses to ethanol. Previous studies on acute ethanol identified genetic variation in brain gene expression networks and behavioral responses to ethanol across the BXD panel of recombinant inbred mice. In this work, we have performed the first joint genetic and genomic analysis of transcriptome shifts in response to chronic intermittent ethanol (CIE) by vapor chamber exposure in a BXD cohort. CIE treatment is known to produce significant and sustained changes in ethanol consumption with repeated cycles of ethanol vapor. Using Affymetrix microarray analysis of prefrontal cortex (PFC) and nucleus accumbens (NAC) RNA, we compared CIE expression responses to those seen following acute ethanol treatment, and to voluntary ethanol consumption. Gene expression changes in PFC and NAC after CIE overlapped significantly across brain regions and with previously published expression following acute ethanol. Genes highly modulated by CIE were enriched for specific biological processes including synaptic transmission, neuron ensheathment, intracellular signaling, and neuronal projection development. Expression quantitative trait locus (eQTL) analyses identified genomic loci associated with ethanol-induced transcriptional changes with largely distinct loci identified between brain regions. Correlating CIE-regulated genes to ethanol consumption data identified specific genes highly associated with variation in the increase in drinking seen with repeated cycles of CIE. In particular, multiple myelin-related genes were identified. Furthermore, genetic variance in or near dynamin3 (Dnm3) on Chr1 at ∼164 Mb may have a major regulatory role in CIE-responsive gene expression. Dnm3 expression correlates significantly with ethanol consumption, is contained in a highly ranked functional group of CIE-regulated genes in the NAC, and has a cis-eQTL within a genomic region linked with multiple CIE-responsive genes.
Collapse
|
15
|
Suryanarayanan A, Carter JM, Landin JD, Morrow AL, Werner DF, Spigelman I. Role of interleukin-10 (IL-10) in regulation of GABAergic transmission and acute response to ethanol. Neuropharmacology 2016; 107:181-188. [PMID: 27016017 PMCID: PMC5076550 DOI: 10.1016/j.neuropharm.2016.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 12/27/2022]
Abstract
Mounting evidence indicates that ethanol (EtOH) exposure activates neuroimmune signaling. Alterations in pro-inflammatory cytokines after acute and chronic EtOH exposure have been heavily investigated. In contrast, little is known about the regulation of neurotransmission and/or modulation by anti-inflammatory cytokines in the brain after an acute EtOH exposure. Recent evidence suggests that interleukin-10 (IL-10), an anti-inflammatory cytokine, is upregulated during withdrawal from chronic EtOH exposure. In the present study, we show that IL-10 is increased early (1 h) after a single intoxicating dose of EtOH (5 g/kg, intragastric) in Sprague Dawley rats. We also show that IL-10 rapidly regulates GABAergic transmission in dentate gyrus neurons. In brain slice recordings, IL-10 application dose-dependently decreases miniature inhibitory postsynaptic current (mIPSC) area and frequency, and decreases the magnitude of the picrotoxin sensitive tonic current (Itonic), indicating both pre- and postsynaptic mechanisms. A PI3K inhibitor LY294002 (but not the negative control LY303511) ablated the inhibitory effects of IL-10 on mIPSC area and Itonic, but not on mIPSC frequency, indicating the involvement of PI3K in postsynaptic effects of IL-10 on GABAergic transmission. Lastly, we also identify a novel neurobehavioral regulation of EtOH sensitivity by IL-10, whereby IL-10 attenuates acute EtOH-induced hypnosis. These results suggest that EtOH causes an early release of IL-10 in the brain, which may contribute to neuronal hyperexcitability as well as disturbed sleep seen after binge exposure to EtOH. These results also identify IL-10 signaling as a potential therapeutic target in alcohol-use disorders and other CNS disorders where GABAergic transmission is altered.
Collapse
Affiliation(s)
- A Suryanarayanan
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia College of Pharmacy, Philadelphia, PA 19104, USA.
| | - J M Carter
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - J D Landin
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - A L Morrow
- Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - D F Werner
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - I Spigelman
- Division of Oral Biology & Medicine, School of Dentistry, 63-078 CHS, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
Carlson SL, Bohnsack JP, Morrow AL. Ethanol Regulation of Synaptic GABAA α4 Receptors Is Prevented by Protein Kinase A Activation. J Pharmacol Exp Ther 2016; 357:10-6. [PMID: 26857960 PMCID: PMC4809316 DOI: 10.1124/jpet.115.230417] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023] Open
Abstract
Ethanol alters GABAA receptor trafficking and function through activation of protein kinases, and these changes may underlie ethanol dependence and withdrawal. In this study, we used subsynaptic fraction techniques and patch-clamp electrophysiology to investigate the biochemical and functional effects of protein kinase A (PKA) and protein kinase C (PKC) activation by ethanol on synaptic GABAA α4 receptors, a key target of ethanol-induced changes. Rat cerebral cortical neurons were grown for 18 days in vitro and exposed to ethanol and/or kinase modulators for 4 hours, a paradigm that recapitulates GABAergic changes found after chronic ethanol exposure in vivo. PKA activation by forskolin or rolipram during ethanol exposure prevented increases in P2 fraction α4 subunit abundance, whereas inhibiting PKA had no effect. Similarly, in the synaptic fraction, activation of PKA by rolipram in the presence of ethanol prevented the increase in synaptic α4 subunit abundance, whereas inhibiting PKA in the presence of ethanol was ineffective. Conversely, PKC inhibition in the presence of ethanol prevented the ethanol-induced increases in synaptic α4 subunit abundance. Finally, we found that either activating PKA or inhibiting PKC in the presence of ethanol prevented the ethanol-induced decrease in GABA miniature inhibitory postsynaptic current decay τ1, whereas inhibiting PKA had no effect. We conclude that PKA and PKC have opposing effects in the regulation of synaptic α4 receptors, with PKA activation negatively modulating, and PKC activation positively modulating, synaptic α4 subunit abundance and function. These results suggest potential targets for restoring normal GABAergic functioning in the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Stephen L Carlson
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - John Peyton Bohnsack
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - A Leslie Morrow
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
17
|
Bohnsack JP, Carlson SL, Morrow AL. Differential regulation of synaptic and extrasynaptic α4 GABA(A) receptor populations by protein kinase A and protein kinase C in cultured cortical neurons. Neuropharmacology 2016; 105:124-132. [PMID: 26767953 DOI: 10.1016/j.neuropharm.2016.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022]
Abstract
The GABAA α4 subunit exists in two distinct populations of GABAA receptors. Synaptic GABAA α4 receptors are localized at the synapse and mediate phasic inhibitory neurotransmission, while extrasynaptic GABAA receptors are located outside of the synapse and mediate tonic inhibitory transmission. These receptors have distinct pharmacological and biophysical properties that contribute to interest in how these different subtypes are regulated under physiological and pathological states. We utilized subcellular fractionation procedures to separate these populations of receptors in order to investigate their regulation by protein kinases in cortical cultured neurons. Protein kinase A (PKA) activation decreases synaptic α4 expression while protein kinase C (PKC) activation increases α4 subunit expression, and these effects are associated with increased β3 S408/409 or γ2 S327 phosphorylation respectively. In contrast, PKA activation increases extrasynaptic α4 and δ subunit expression, while PKC activation has no effect. Our findings suggest synaptic and extrasynaptic GABAA α4 subunit expression can be modulated by PKA to inform the development of more specific therapeutics for neurological diseases that involve deficits in GABAergic transmission.
Collapse
Affiliation(s)
- John Peyton Bohnsack
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - Stephen L Carlson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - A Leslie Morrow
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA.
| |
Collapse
|
18
|
Carlson SL, Bohnsack JP, Patel V, Morrow AL. Regulation of Extrasynaptic GABAA α4 Receptors by Ethanol-Induced Protein Kinase A, but Not Protein Kinase C Activation in Cultured Rat Cerebral Cortical Neurons. J Pharmacol Exp Ther 2016; 356:148-56. [PMID: 26483396 PMCID: PMC4702069 DOI: 10.1124/jpet.115.228056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/14/2015] [Indexed: 01/14/2023] Open
Abstract
Ethanol produces changes in GABAA receptor trafficking and function that contribute to ethanol dependence symptomatology. Extrasynaptic γ-aminobutyric acid A receptors (GABAA-R) mediate inhibitory tonic current and are of particular interest because they are potentiated by physiologically relevant doses of ethanol. Here, we isolate GABAA α4δ receptors by western blotting in subsynaptic fractions to investigate protein kinase A (PKA) and protein kinase C (PKC) modulation of ethanol-induced receptor trafficking, while extrasynaptic receptor function is determined by measurement of tonic inhibition and responses evoked by 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol (THIP). Rat cerebral cortical neurons were grown for 18 days in vitro and exposed to ethanol and/or PKA/PKC modulators. Ethanol exposure (1 hour) did not alter GABAA α4 receptor abundance, but it increased tonic current amplitude, an effect that was prevented by inhibiting PKA, but not PKC. Direct activation of PKA, but not PKC, increased the abundance and tonic current of extrasynaptic α4δ receptors. In contrast, prolonged ethanol exposure (4 hours) reduced α4δ receptor abundance as well as tonic current, and this effect was also PKA dependent. Finally, PKC activation by ethanol or phorbol-12,13-dibutyrate (PdBu) had no effect on extrasynaptic α4δ subunit abundance or activity. We conclude that ethanol alters extrasynaptic α4δ receptor function and expression in cortical neurons in a PKA-dependent manner, but ethanol activation of PKC does not influence these receptors. These results could have clinical relevance for therapeutic strategies to restore normal GABAergic functioning for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Stephen L Carlson
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - J Peyton Bohnsack
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Vraj Patel
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - A Leslie Morrow
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
19
|
Robinson JE, Vardy E, DiBerto JF, Chefer VI, White KL, Fish EW, Chen M, Gigante E, Krouse MC, Sun H, Thorsell A, Roth BL, Heilig M, Malanga CJ. Receptor Reserve Moderates Mesolimbic Responses to Opioids in a Humanized Mouse Model of the OPRM1 A118G Polymorphism. Neuropsychopharmacology 2015; 40:2614-22. [PMID: 25881115 PMCID: PMC4569952 DOI: 10.1038/npp.2015.109] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/09/2022]
Abstract
The OPRM1 A118G polymorphism is the most widely studied μ-opioid receptor (MOR) variant. Although its involvement in acute alcohol effects is well characterized, less is known about the extent to which it alters responses to opioids. Prior work has shown that both electrophysiological and analgesic responses to morphine but not to fentanyl are moderated by OPRM1 A118G variation, but the mechanism behind this dissociation is not known. Here we found that humanized mice carrying the 118GG allele (h/mOPRM1-118GG) were less sensitive than h/mOPRM1-118AA littermates to the rewarding effects of morphine and hydrocodone but not those of other opioids measured with intracranial self-stimulation. Reduced morphine reward in 118GG mice was associated with decreased dopamine release in the nucleus accumbens and reduced effects on GABA release in the ventral tegmental area that were not due to changes in drug potency or efficacy in vitro or receptor-binding affinity. Fewer MOR-binding sites were observed in h/mOPRM1-118GG mice, and pharmacological reduction of MOR availability unmasked genotypic differences in fentanyl sensitivity. These findings suggest that the OPRM1 A118G polymorphism decreases sensitivity to low-potency agonists by decreasing receptor reserve without significantly altering receptor function.
Collapse
Affiliation(s)
- J Elliott Robinson
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eyal Vardy
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeffrey F DiBerto
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Vladimir I Chefer
- Intramural Research Program, National Institute on Drug Abuse (NIDA), Baltimore, MD, USA
| | - Kate L White
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric W Fish
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Meng Chen
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eduardo Gigante
- Intramural Research Program, National Institute on Drug Abuse (NIDA), Baltimore, MD, USA
| | - Michael C Krouse
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hui Sun
- Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, USA
| | - Annika Thorsell
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,NIMH Psychoactive Drug Screening Program (PDSP), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Markus Heilig
- Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, USA,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C J Malanga
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Department of Neurology, University of North Carolina School of Medicine, Physicians' Office Building, 170 Manning Drive, CB 7025, Chapel Hill, NC 27599-7025, USA, Tel: +1 919 966 1683, Fax: +1 919 843 4576, E-mail:
| |
Collapse
|
20
|
Zhao W, Wang J, Song S, Li F, Yuan F. Reduction of α1GABAA receptor mediated by tyrosine kinase C (PKC) phosphorylation in a mouse model of fragile X syndrome. Int J Clin Exp Med 2015; 8:13219-26. [PMID: 26550246 PMCID: PMC4612931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/11/2015] [Indexed: 06/05/2023]
Abstract
Fragile X syndrome (FXS) caused by lack of fragile X mental retardation protein (Fmr1) is the most common cause of inherited intellectual disability and characterized by many cognitive disturbances like attention deficit, autistic behavior, and audiogenic seizure and have region-specific altered expression of some gamma-aminobutyric acid (GABAA) receptor subunits. Quantitative real-time polymerase chain reaction and western blot experiments were performed in the cultured cortical neurons and forebrain obtained from wild-type (WT) and Fmr1 KO mice demonstrate the reduction in the expression of α1 gamma-aminobutyric acid (α1GABAA) receptor, phospho-α1GABAA receptor, PKC and phosphor-PKC in Fmr1 KO mice comparing with WT mice, both in vivo and in vitro. Furthermore, we found that the phosphorylation of the α1GABAA receptor was mediated by PKC. Our results elucidate that the lower phosphorylation of the α1GABAA receptor mediated by PKC neutralizes the seizure-promoting effects in Fmr1 KO mice and point to the potential therapeutic targets of α1GABAA agonists for the treatment of fragile X syndrome.
Collapse
Affiliation(s)
- Weidong Zhao
- Department of Habilitation, The Central Hospital of Xinxiang CityXinxiang, Henan Province, P. R. China
| | - Jiaqin Wang
- Department of Pediatrics, The Third Affiliated Hospital of Xinxiang Medical CollegeHenan Province, P. R. China
| | - Shunyi Song
- Department of Habilitation, The Central Hospital of Xinxiang CityXinxiang, Henan Province, P. R. China
| | - Fang Li
- Department of Habilitation, The Central Hospital of Xinxiang CityXinxiang, Henan Province, P. R. China
| | - Fangfang Yuan
- Department of Habilitation, The Central Hospital of Xinxiang CityXinxiang, Henan Province, P. R. China
| |
Collapse
|
21
|
Nakamura Y, Darnieder LM, Deeb TZ, Moss SJ. Regulation of GABAARs by phosphorylation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 72:97-146. [PMID: 25600368 PMCID: PMC5337123 DOI: 10.1016/bs.apha.2014.11.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are the principal mediators of fast synaptic inhibition in the brain as well as the low persistent extrasynaptic inhibition, both of which are fundamental to proper brain function. Thus unsurprisingly, deficits in GABAARs are implicated in a number of neurological disorders and diseases. The complexity of GABAAR regulation is determined not only by the heterogeneity of these receptors but also by its posttranslational modifications, the foremost, and best characterized of which is phosphorylation. This review will explore the details of this dynamic process, our understanding of which has barely scratched the surface. GABAARs are regulated by a number of kinases and phosphatases, and its phosphorylation plays an important role in governing its trafficking, expression, and interaction partners. Here, we summarize the progress in understanding the role phosphorylation plays in the regulation of GABAARs. This includes how phosphorylation can affect the allosteric modulation of GABAARs, as well as signaling pathways that affect GABAAR phosphorylation. Finally, we discuss the dysregulation of GABAAR phosphorylation and its implication in disease processes.
Collapse
|
22
|
Wang S, Luo Y, Feng A, Li T, Yang X, Nofech-Mozes R, Yu M, Wang C, Li Z, Yi F, Liu C, Lu WY. Ethanol induced impairment of glucose metabolism involves alterations of GABAergic signaling in pancreatic β-cells. Toxicology 2014; 326:44-52. [PMID: 25456265 DOI: 10.1016/j.tox.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 01/02/2023]
Abstract
Alcohol overindulgence is a risk factor of type 2 diabetes mellitus. However, the mechanisms by which alcohol overindulgence damages glucose metabolism remain unclear. Pancreatic islet β-cells are endowed with type-A γ-aminobutyric acid receptor (GABAAR) mediated autocrine signaling mechanism, which regulates insulin secretion and fine-tunes glucose metabolism. In neurons GABAAR is one of the major targets for alcohol. This study investigated whether ethanol alters glucose metabolism by affecting GABAAR signaling in pancreatic β-cells. Blood glucose level of test mice was measured using a blood glucose meter. Insulin secretion by the pancreatic β-cell line INS-1 cells was examined using a specific insulin ELISA kit. Whole-cell patch-clamp recording was used to evaluate GABA-elicited current in INS-1 cells. Western blot and immunostaining were used to measure the expression of GABAAR subunits in mouse pancreatic tissues or in INS-1 cells. Intraperitoneal (i.p.) administration of ethanol (3.0g/kg body weight) to mice altered glucose metabolism, which was associated with decreased expression of GABAAR α1- and δ- subunits on the surface of pancreatic β-cells. Acute treatment of cultured INS-1cells with ethanol (60mM) decreased the GABA-induced current and reduced insulin secretion. In contrast, treating INS-1 cells with GABA (100μM) largely prevented the ethanol-induced reduction of insulin release. Importantly, pre-treating mice with GABA (i.p., 1.5mg/kg body weight) partially reversed ethanol-induced impairment of glucose homeostasis in mice. Our data suggest a novel role of pancreatic GABA signaling in protecting pancreatic islet β-cells from ethanol-induced dysfunction.
Collapse
Affiliation(s)
- Shuanglian Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Yan Luo
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Allen Feng
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Tao Li
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xupeng Yang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Roy Nofech-Mozes
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Meng Yu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Changhui Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Ziwei Li
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Chuanyong Liu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China.
| | - Wei-Yang Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
23
|
Liang J, Lindemeyer AK, Suryanarayanan A, Meyer EM, Marty VN, Ahmad SO, Shao XM, Olsen RW, Spigelman I. Plasticity of GABA(A) receptor-mediated neurotransmission in the nucleus accumbens of alcohol-dependent rats. J Neurophysiol 2014; 112:39-50. [PMID: 24694935 DOI: 10.1152/jn.00565.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chronic alcohol exposure-induced changes in reinforcement mechanisms and motivational state are thought to contribute to the development of cravings and relapse during protracted withdrawal. The nucleus accumbens (NAcc) is a key structure of the mesolimbic dopaminergic reward system and plays an important role in mediating alcohol-seeking behaviors. Here we describe the long-lasting alterations of γ-aminobutyric acid type A receptors (GABA(A)Rs) of medium spiny neurons (MSNs) in the NAcc after chronic intermittent ethanol (CIE) treatment, a rat model of alcohol dependence. CIE treatment and withdrawal (>40 days) produced decreases in the ethanol and Ro15-4513 potentiation of extrasynaptic GABA(A)Rs, which mediate the picrotoxin-sensitive tonic current (I(tonic)), while potentiation of synaptic receptors, which give rise to miniature inhibitory postsynaptic currents (mIPSCs), was increased. Diazepam sensitivity of both I(tonic) and mIPSCs was decreased by CIE treatment. The average magnitude of I(tonic) was unchanged, but mIPSC amplitude and frequency decreased and mIPSC rise time increased after CIE treatment. Rise-time histograms revealed decreased frequency of fast-rising mIPSCs after CIE treatment, consistent with possible decreases in somatic GABAergic synapses in MSNs from CIE rats. However, unbiased stereological analysis of NeuN-stained NAcc neurons did not detect any decreases in NAcc volume, neuronal numbers, or neuronal cell body volume. Western blot analysis of surface subunit levels revealed selective decreases in α1 and δ and increases in α4, α5, and γ2 GABA(A)R subunits after CIE treatment and withdrawal. Similar, but reversible, alterations occurred after a single ethanol dose (5 g/kg). These data reveal CIE-induced long-lasting neuroadaptations in the NAcc GABAergic neurotransmission.
Collapse
Affiliation(s)
- Jing Liang
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - A Kerstin Lindemeyer
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Asha Suryanarayanan
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California
| | - Edward M Meyer
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California
| | - Vincent N Marty
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California
| | - S Omar Ahmad
- Doisy College of Health Sciences, Saint Louis University, St. Louis, Missouri; and
| | - Xuesi Max Shao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Richard W Olsen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Igor Spigelman
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California;
| |
Collapse
|
24
|
Santerre JL, Gigante ED, Landin JD, Werner DF. Molecular and behavioral characterization of adolescent protein kinase C following high dose ethanol exposure. Psychopharmacology (Berl) 2014; 231:1809-20. [PMID: 24051603 PMCID: PMC4012395 DOI: 10.1007/s00213-013-3267-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/20/2013] [Indexed: 11/26/2022]
Abstract
RATIONALE Ethanol is commonly used and abused during adolescence. Although adolescents display differential behavioral responses to ethanol, the mechanisms by which this occurs are not known. The protein kinase C (PKC) pathway has been implicated in mediating many ethanol-related effects in adults, as well as gamma-aminobutyric acid (GABA(A)) receptor regulation. OBJECTIVES The present study was designed to characterize cortical PKC isoform and GABA(A) receptor subunit expression during adolescence relative to adults as well as assess PKC involvement in ethanol action. RESULTS Novel PKC isoforms were elevated, while PKCγ was lower during mid-adolescence relative to adults. Whole-cell lysate and synaptosomal preparations correlated for all isoforms except PKCδ. In parallel, synaptosomal GABAA receptor subunit expression was also developmentally regulated, with GABA(A)R δ and α4 being lower while α1 and γ2 were higher or similar, respectively, in adolescents compared to adults. Following acute ethanol exposure, synaptosomal novel and atypical PKC isoform expression was decreased only in adolescents. Behaviorally, inhibiting PKC with calphostin C, significantly increased ethanol-induced loss of righting reflex (LORR) in adolescents but not adults, whereas activating PKC with phorbol dibutyrate was ineffective in adolescents but decreased LORR duration in adults. Further investigation revealed that inhibiting the cytosolic phospholipase A2/arachidonic acid (cPLA2/AA) pathway increased LORR duration in adolescents, but was ineffective in adults. CONCLUSIONS These data indicate that PKC isoforms are variably regulated during adolescence and may contribute to adolescent ethanol-related behavior. Furthermore, age-related differences in the cPLA2/AA pathway may contribute to ethanol's age-related effects on novel and atypical PKC isoform expression and behavior.
Collapse
Affiliation(s)
- Jessica L. Santerre
- Department of Psychology, Binghamton University, Binghamton, New York
- Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, New York
| | - Eduardo D. Gigante
- Department of Psychology, Binghamton University, Binghamton, New York
- Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, New York
| | - Justine D. Landin
- Department of Psychology, Binghamton University, Binghamton, New York
- Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, New York
| | - David F. Werner
- Department of Psychology, Binghamton University, Binghamton, New York
- Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, New York
| |
Collapse
|
25
|
Tabakoff B, Hoffman PL. The neurobiology of alcohol consumption and alcoholism: an integrative history. Pharmacol Biochem Behav 2013; 113:20-37. [PMID: 24141171 PMCID: PMC3867277 DOI: 10.1016/j.pbb.2013.10.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 01/17/2023]
Abstract
Studies of the neurobiological predisposition to consume alcohol (ethanol) and to transition to uncontrolled drinking behavior (alcoholism), as well as studies of the effects of alcohol on brain function, started a logarithmic growth phase after the repeal of the 18th Amendment to the United States Constitution. Although the early studies were primitive by current technological standards, they clearly demonstrated the effects of alcohol on brain structure and function, and by the end of the 20th century left little doubt that alcoholism is a "disease" of the brain. This review traces the history of developments in the understanding of ethanol's effects on the most prominent inhibitory and excitatory systems of brain (GABA and glutamate neurotransmission). This neurobiological information is integrated with knowledge of ethanol's actions on other neurotransmitter systems to produce an anatomical and functional map of ethanol's properties. Our intent is limited in scope, but is meant to provide context and integration of the actions of ethanol on the major neurobiologic systems which produce reinforcement for alcohol consumption and changes in brain chemistry that lead to addiction. The developmental history of neurobehavioral theories of the transition from alcohol drinking to alcohol addiction is presented and juxtaposed to the neurobiological findings. Depending on one's point of view, we may, at this point in history, know more, or less, than we think we know about the neurobiology of alcoholism.
Collapse
Affiliation(s)
- Boris Tabakoff
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| | - Paula L. Hoffman
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| |
Collapse
|
26
|
Carlson SL, O'Buckley TK, Thomas R, Thiele TE, Morrow AL. Altered GABAA receptor expression and seizure threshold following acute ethanol challenge in mice lacking the RIIβ subunit of PKA. Neurochem Res 2013; 39:1079-87. [PMID: 24104609 DOI: 10.1007/s11064-013-1167-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/25/2013] [Accepted: 09/27/2013] [Indexed: 11/26/2022]
Abstract
Ethanol causes pathological changes in GABAA receptor trafficking and function. These changes are mediated in part by ethanol activation of protein kinase A (PKA). The current study investigated the expression of the GABAA α1 and α4 subunits and the kinase anchoring protein AKAP150, as well as bicuculline-induced seizure threshold, at baseline and following acute injection of ethanol (3.5 g/kg IP) in a mouse line lacking the regulatory RIIβ subunit of PKA. Whole cerebral cortices were harvested at baseline, 1 h, or 46 h following injection of ethanol or saline and subjected to fractionation and western blot analysis. Knockout (RIIβ-/-) mice had similar baseline levels of PKA RIIα and GABAA α1 and α4 subunits compared to wild type (RIIβ+/+) littermates, but had deficits in AKAP150. GABAA α1 subunit levels were decreased in the P2 fraction of RIIβ-/-, but not RIIβ+/+, mice following 1 h ethanol, an effect that was driven by decreased α1 expression in the synaptic fraction. GABAA α4 subunits in the P2 fraction were not affected by 1 h ethanol; however, synaptic α4 subunit expression was increased in RIIβ+/+, but not RIIβ-/- mice, while extrasynaptic α4 and δ subunit expression were decreased in RIIβ-/-, but not RIIβ+/+ mice. Finally, RIIβ knockout was protective against bicuculline-induced seizure susceptibility. Overall, the results suggest that PKA has differential roles in regulating GABAA receptor subunits. PKA may protect against ethanol-induced deficits in synaptic α1 and extrasynaptic α4 receptors, but may facilitate the increase of synaptic α4 receptors.
Collapse
Affiliation(s)
- Stephen L Carlson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | |
Collapse
|
27
|
Carlson SL, Kumar S, Werner DF, Comerford CE, Morrow AL. Ethanol activation of protein kinase A regulates GABAA α1 receptor function and trafficking in cultured cerebral cortical neurons. J Pharmacol Exp Ther 2013; 345:317-25. [PMID: 23408117 PMCID: PMC3629799 DOI: 10.1124/jpet.112.201954] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/12/2013] [Indexed: 12/24/2022] Open
Abstract
Ethanol exposure produces alterations in GABAergic signaling that are associated with dependence and withdrawal. Previously, we demonstrated that ethanol-induced protein kinase C (PKC) γ signaling selectively contributes to changes in GABAA α1 synaptic receptor activity and surface expression. Here, we demonstrate that protein kinase A (PKA) exerts opposing effects on GABAA receptor adaptations during brief ethanol exposure. Cerebral cortical neurons from day 0-1 rat pups were tested after 18 days in culture. Receptor trafficking was assessed by Western blot analysis, and functional changes were measured using whole-cell patch-clamp recordings of evoked and miniature inhibitory postsynaptic current (mIPSC) responses. One-hour ethanol exposure increased membrane-associated PKC and PKA, but steady-state GABAA α1 subunit levels were maintained. Activation of PKA by Sp-adenosine 3',5'-cyclic monophosphothioate triethylamine alone increased GABAA α1 subunit surface expression and zolpidem potentiation of GABA responses, whereas coexposure of ethanol with the PKA inhibitor Rp-adenosine 3',5'-cyclic monophosphothioate triethylamine decreased α1 subunit expression and zolpidem responses. Exposure to the PKC inhibitor calphostin-C with ethanol mimicked the effect of direct PKA activation. The effects of PKA modulation on mIPSC decay τ were consistent with its effects on GABA currents evoked in the presence of zolpidem. Overall, the results suggest that PKA acts in opposition to PKC on α1-containing GABAA receptors, mediating the GABAergic effects of ethanol exposure, and may provide an important target for the treatment of alcohol dependence/withdrawal.
Collapse
Affiliation(s)
- Stephen L Carlson
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
28
|
Ethanol promotes clathrin adaptor-mediated endocytosis via the intracellular domain of δ-containing GABAA receptors. J Neurosci 2013; 32:17874-81. [PMID: 23223306 DOI: 10.1523/jneurosci.2535-12.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Pharmacological and genetic evidence reveals that GABA(A) receptor (GABA(A)-R) expression and localization are modulated in response to acute and chronic ethanol (EtOH) exposure. To determine molecular mechanisms of GABA(A)-R plasticity in response to in vivo acute EtOH, we measured early time changes in GABA(A)-R subunit localization. Single doses of EtOH (3 g/kg via i.p. injection in rats) produced decreases in surface levels of GABA(A)-R α4 and δ subunits at 5-15 min post-EtOH in hippocampus CA1 and dentate gyrus, verifying our earlier report (Liang et al., 2007). Here we also examined the β3 subunit and its phosphorylation state during internalization. β3 also was internalized during 5-15 min after EtOH exposure, while phosphorylation of β3 was increased, then decreased at later times, ruling out β3 dephosphorylation-dependent endocytosis. As early as 5 min post-EtOH, there is an initial increase in association between the δ subunits with clathrin adaptor proteins AP2-μ2 revealed by coimmunoprecipitation, followed by a decrease in association 15 min post-EtOH. In vitro studies using glutathione S-transferase fused to the δ subunit intracellular domain (ICD) show that two regions, one containing a classical YxxΦ motif and the other an atypical R/K-rich motif, directly and differentially bind to AP2-μ2, with the former YRSV exhibiting higher affinity. Mutating both regions in the δ-ICD abolishes μ2 binding, providing a possible mechanism that can explain the rapid downregulation of extrasynaptic α4βδ-GABA(A)-R following in vivo EtOH administration, in which the δ-ICD increases in affinity for clathrin AP2-μ2 leading to endocytosis.
Collapse
|
29
|
Shan HQ, Hammarback JA, Godwin DW. Ethanol inhibition of a T-type Ca²+ channel through activity of protein kinase C. Alcohol Clin Exp Res 2013; 37:1333-42. [PMID: 23488970 DOI: 10.1111/acer.12098] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 01/02/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND T-type calcium channels (T-channels) are widely distributed in the central and peripheral nervous system, where they mediate calcium entry and regulate the intrinsic excitability of neurons. T-channels are dysregulated in response to alcohol administration and withdrawal. We therefore investigated acute ethanol (EtOH) effects and the underlying mechanism of action in human embryonic kidney (HEK) 293 cell lines, as well as effects on native currents recorded from dorsal root ganglion (DRG) neurons cultured from Long-Evans rats. METHODS Whole-cell voltage-clamp recordings were performed at 32 to 34°C in both HEK cell lines and DRG neurons. The recordings were taken after a 10-minute application of EtOH or protein kinase C (PKC) activator (phorbol 12-myristate 13-acetate [PMA]). RESULTS We recorded T-type Ca²⁺ currents (T-currents) from 3 channel isoforms (CaV3.1, CaV3.2, and CaV3.3) before and during administration of EtOH. We found that only 1 isoform, CaV3.2, was significantly affected by EtOH. EtOH reduced current density as well as producing a hyperpolarizing shift in steady-state inactivation of both CaV3.2 currents from HEK 293 cell lines and in native T-currents from DRG neurons that are known to be enriched in CaV3.2. A myristoylated PKC peptide inhibitor (MPI) blocked the major EtOH effects, in both the cell lines and the DRG neurons. However, PMA effects were more complex. Lower concentration PMA (100 nM) replicated the major effects of EtOH, while higher concentration PMA (1 μM) did not, suggesting that the EtOH effects operate through activation of PKC and were mimicked by lower concentration of PMA. CONCLUSIONS EtOH primarily affects the CaV3.2 isoform of T-type Ca²⁺ channels acting through PKC, highlighting a novel target and mechanism for EtOH effects on excitable membranes.
Collapse
Affiliation(s)
- Hong Qu Shan
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | |
Collapse
|
30
|
Wang Y, Wu J, Guo R, Zhao Y, Wang Y, Zhang M, Chen Z, Wu A, Yue Y. Surgical incision induces phosphorylation of AMPA receptor GluR1 subunits at Serine-831 sites and GluR1 trafficking in spinal cord dorsal horn via a protein kinase Cγ-dependent mechanism. Neuroscience 2013; 240:361-70. [PMID: 23470774 DOI: 10.1016/j.neuroscience.2013.02.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/14/2013] [Accepted: 02/25/2013] [Indexed: 01/07/2023]
Abstract
Spinal α-amino-3-hydroxy-5-methy-4-isoxazole propionate (AMPA) receptor plays an important role in acute pain induced by surgical tissue injuries. Our previous study has shown that the enhanced phosphorylation of AMPA receptor GluR1 subunits at Serine-831 sites by protein kinase C (PKC) in the spinal cord dorsal horn is involved in post-surgical pain hypersensitivity. However, which isoforms of PKC are responsible for the phosphorylation of AMPA receptor GluR1 subunits at Serine-831 sites remains to be established. In the present study, using an animal model of postoperative pain, we found that surgical tissue injuries enhanced the membrane translocation level of PKCγ, but not PKCα, βI, and βII, and induced the trafficking of GluR1, but not GluR2 into neuronal plasma membrane. Intrathecal (i.t.) pretreatment of small interfering RNA targeting PKCγ to reduce the PKCγ expression in the spinal cord significantly attenuated the pain hypersensitivity and inhibited the phosphorylation of AMPA receptor GluR1 subunits at Serine-831 sites as well as GluR1 membrane trafficking. Our study indicates that the surgical incision-induced phosphorylation of AMPA receptor GluR1 subunits at Serine-831 sites and GluR1 trafficking are regulated by a PKCγ-dependent mechanism.
Collapse
Affiliation(s)
- Y Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Ethanol's effects on intracellular signaling pathways contribute to acute effects of ethanol as well as to neuroadaptive responses to repeated ethanol exposure. In this chapter we review recent discoveries that demonstrate how ethanol alters signaling pathways involving several receptor tyrosine kinases and intracellular tyrosine and serine-threonine kinases, with consequences for regulation of cell surface receptor function, gene expression, protein translation, neuronal excitability and animal behavior. We also describe recent work that demonstrates a key role for ethanol in regulating the function of scaffolding proteins that organize signaling complexes into functional units. Finally, we review recent exciting studies demonstrating ethanol modulation of DNA and histone modification and the expression of microRNAs, indicating epigenetic mechanisms by which ethanol regulates neuronal gene expression and addictive behaviors.
Collapse
Affiliation(s)
- Dorit Ron
- Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Robert O. Messing
- Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| |
Collapse
|
32
|
Zeng L, Webster SV, Newton PM. The biology of protein kinase C. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:639-61. [PMID: 22453963 DOI: 10.1007/978-94-007-2888-2_28] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review gives a basic introduction to the biology of protein kinase C, one of the first calcium-dependent kinases to be discovered. We review the structure and function of protein kinase C, along with some of the substrates of individual isoforms. We then review strategies for inhibiting PKC in experimental systems and finally discuss the therapeutic potential of targeting PKC. Each aspect is covered in summary, with links to detailed resources where appropriate.
Collapse
Affiliation(s)
- Lily Zeng
- School of Medicine, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
33
|
Mattoo SK, Gaur N, Das PP. Zolpidem withdrawal delirium. Indian J Pharmacol 2012; 43:729-30. [PMID: 22144786 PMCID: PMC3229797 DOI: 10.4103/0253-7613.89838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 04/08/2011] [Accepted: 08/31/2011] [Indexed: 01/24/2023] Open
Abstract
The Z-category hypnotics are promoted for their relative safety. However, this view is challenged by the emerging clinical evidence in the form of zolpidem related intoxication delirium and seizures, and dependence and complicated withdrawal. We report the case of a zolpidem-naive alcohol-dependent inpatient that, while undergoing alcohol de-addiction, was prescribed zolpidem for insomnia and developed delirium during taper-off. He was successfully detoxified for alcohol, treated for delirium and put on disulfiram prophylaxis. The case highlights the need for being cautious while using zolpidem for insomnia in alcohol dependent subjects.
Collapse
Affiliation(s)
- Surendra K Mattoo
- Department of Psychiatry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
34
|
Kumar S, Ren Q, Beckley JH, O'Buckley TK, Gigante ED, Santerre JL, Werner DF, Morrow AL. Ethanol Activation of Protein Kinase A Regulates GABA(A) Receptor Subunit Expression in the Cerebral Cortex and Contributes to Ethanol-Induced Hypnosis. Front Neurosci 2012; 6:44. [PMID: 22509146 PMCID: PMC3321501 DOI: 10.3389/fnins.2012.00044] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 03/19/2012] [Indexed: 11/13/2022] Open
Abstract
Protein kinases are implicated in neuronal cell functions such as modulation of ion channel function, trafficking, and synaptic excitability. Both protein kinase C (PKC) and A (PKA) are involved in regulation of γ-aminobutyric acid type A (GABAA) receptors through phosphorylation. However, the role of PKA in regulating GABAA receptors (GABAA-R) following acute ethanol exposure is not known. The present study investigated the role of PKA in the effects of ethanol on GABAA-R α1 subunit expression in rat cerebral cortical P2 synaptosomal fractions. Additionally, GABA-related behaviors were examined. Rats were administered ethanol (2.0–3.5 g/kg) or saline and PKC, PKA, and GABAA-R α1 subunit levels were measured by western blot analysis. Ethanol (3.5 g/kg) transiently increased GABAA-R α1 subunit expression and PKA RIIβ subunit expression at similar time points whereas PKA RIIα was increased at later time points. In contrast, PKC isoform expression remained unchanged. Notably, lower ethanol doses (2.0 g/kg) had no effect on GABAA-R α1 subunit levels, although PKA type II regulatory subunits RIIα and RIIβ were increased at 10 and 60 min when PKC isozymes are also known to be elevated. To determine if PKA activation was responsible for the ethanol-induced elevation of GABAA-R α1 subunits, the PKA antagonist H89 was administered to rats prior to ethanol exposure. H89 administration prevented ethanol-induced increases in GABAA-R α1 subunit expression. Moreover, increasing PKA activity intracerebroventricularly with Sp-cAMP prior to a hypnotic dose of ethanol increased ethanol-induced loss of righting reflex (LORR) duration. This effect appears to be mediated in part by GABAA-R as increasing PKA activity also increased the duration of muscimol-induced LORR. Overall, these data suggest that PKA mediates ethanol-induced GABAA-R expression and contributes to behavioral effects of ethanol involving GABAA-R.
Collapse
Affiliation(s)
- Sandeep Kumar
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine Chapel Hill, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
McCool BA. Ethanol modulation of synaptic plasticity. Neuropharmacology 2011; 61:1097-108. [PMID: 21195719 PMCID: PMC3149748 DOI: 10.1016/j.neuropharm.2010.12.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/10/2010] [Accepted: 12/22/2010] [Indexed: 12/19/2022]
Abstract
Synaptic plasticity in the most general terms represents the flexibility of neurotransmission in response to neuronal activity. Synaptic plasticity is essential both for the moment-by-moment modulation of neural activity in response to dynamic environmental cues and for long-term learning and memory formation. These temporal characteristics are served by an array of pre- and post-synaptic mechanisms that are frequently modulated by ethanol exposure. This modulation likely makes significant contributions to both alcohol abuse and dependence. In this review, I discuss the modulation of both short-term and long-term synaptic plasticity in the context of specific ethanol-sensitive cellular substrates. A general discussion of the available preclinical, animal-model based neurophysiology literature provides a comparison between results from in vitro and in vivo studies. Finally, in the context of alcohol abuse and dependence, the review proposes potential behavioral contributions by ethanol modulation of plasticity.
Collapse
Affiliation(s)
- Brian A McCool
- Department of Physiology & Pharmacology and the Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest University School of Medicine, Winston-Salem NC 27157, USA.
| |
Collapse
|
36
|
Velázquez-Flores MÁ, Salceda R. Glycine receptor internalization by protein kinases activation. Synapse 2011; 65:1231-8. [PMID: 21656573 DOI: 10.1002/syn.20963] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 05/27/2011] [Accepted: 05/31/2011] [Indexed: 11/06/2022]
Abstract
Although glycine-induced currents in the central nervous system have been proven to be modulated by protein kinases A (PKA) and C (PKC), the mechanism is not well understood. In order to better comprehend the mechanism involved in this phenomenon, we tested the PKA and PKC activation effect on the specific [(3) H]glycine and [(3) H]strychnine binding to postsynaptic glycine receptor (GlyR) in intact rat retina. The specific binding constituted about 20% of the total radioligand binding. Kinetic analysis of the specific binding exhibited a sigmoidal behavior with three glycine and two strychnine binding sites and affinities of 212 nM for [(3) H]glycine and 50 nM for [(3) H]strychnine. Specific radioligand binding was decreased (60-85%) by PKA and PKC activation, an effect that was blocked by specific kinases inhibitors, as well as by cytochalasin D. GlyR expressed in the plasma membrane decreased about 50% in response to kinases activation, which was consistent with an increase of the receptor in the microsomal fraction when PKA was activated. Moreover, immunoprecipitation studies indicated that these kinases lead to a time-dependent receptor phosphorylation. Our results suggest that in retina, GlyR is cross-regulated by G protein-coupled receptors, activating PKA and PKC.
Collapse
Affiliation(s)
- Miguel Ángel Velázquez-Flores
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 México, D.F., México.
| | | |
Collapse
|
37
|
Shen Y, Lindemeyer AK, Spigelman I, Sieghart W, Olsen RW, Liang J. Plasticity of GABAA receptors after ethanol pre-exposure in cultured hippocampal neurons. Mol Pharmacol 2011; 79:432-42. [PMID: 21163967 PMCID: PMC3061361 DOI: 10.1124/mol.110.068650] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 12/15/2010] [Indexed: 12/23/2022] Open
Abstract
Alcohol use causes many physiological changes in brain with behavioral sequelae. We previously observed (J Neurosci 27:12367-12377, 2007) plastic changes in hippocampal slice recordings paralleling behavioral changes in rats treated with a single intoxicating dose of ethanol (EtOH). Here, we were able to reproduce in primary cultured hippocampal neurons many of the effects of in vivo EtOH exposure on GABA(A) receptors (GABA(A)Rs). Cells grown 11 to 15 days in vitro demonstrated GABA(A)R δ subunit expression and sensitivity to enhancement by short-term exposure to EtOH (60 mM) of GABA(A)R-mediated tonic current (I(tonic)) using whole-cell patch-clamp techniques. EtOH gave virtually no enhancement of mIPSCs. Cells pre-exposed to EtOH (60 mM) for 30 min showed, 1 h after EtOH withdrawal, a 50% decrease in basal I(tonic) magnitude and tolerance to short-term EtOH enhancement of I(tonic), followed by reduced basal mIPSC area at 4 h. At 24 h, we saw considerable recovery in mIPSC area and significant potentiation by short-term EtOH; in addition, GABA(A)R currents exhibited reduced enhancement by benzodiazepines. These changes paralleled significant decreases in cell-surface expression of normally extrasynaptic δ and α4 GABA(A)R subunits as early as 20 min after EtOH exposure and reduced α5-containing GABA(A)Rs at 1 h, followed by a larger reduction of normally synaptic α1 subunit at 4 h, and then by increases in α4γ2-containing cell-surface receptors by 24 h. Measuring internalization of biotinylated GABA(A)Rs, we showed for the first time that the EtOH-induced loss of I(tonic) and cell-surface δ/α4 20 min after withdrawal results from increased receptor endocytosis rather than decreased exocytosis.
Collapse
Affiliation(s)
- Yi Shen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1735, USA
| | | | | | | | | | | |
Collapse
|
38
|
Werner DF, Kumar S, Criswell HE, Suryanarayanan A, Fetzer JA, Comerford CE, Morrow AL. PKCγ is required for ethanol-induced increases in GABA(A) receptor α4 subunit expression in cultured cerebral cortical neurons. J Neurochem 2011; 116:554-63. [PMID: 21155805 PMCID: PMC3033448 DOI: 10.1111/j.1471-4159.2010.07140.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ethanol exposure produces alterations in GABA(A) receptor function and expression associated with CNS hyperexcitability, but the mechanisms of these effects are unknown. Ethanol is known to increase both GABA(A) receptor α4 subunits and protein kinase C (PKC) isozymes in vivo and in vitro. Here, we investigated ethanol regulation of GABA(A) receptor α4 subunit expression in cultured cortical neurons to delineate the role of PKC. Cultured neurons were prepared from rat pups on postnatal day 0-1 and tested after 18 days. GABA(A) receptor α4 subunit surface expression was assessed using P2 fractionation and surface biotinylation following ethanol exposure for 4 h. Miniature inhibitory post-synaptic currents were measured using whole cell patch clamp recordings. Ethanol increased GABA(A) receptor α4 subunit expression in both the P2 and biotinylated fractions, while reducing the decay time constant in miniature inhibitory post-synaptic currents, with no effect on γ2 or δ subunits. PKC activation mimicked ethanol effects, while the PKC inhibitor calphostin C prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression. PKCγ siRNA knockdown prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression, but inhibition of the PKCβ isoform with PKCβ pseudosubstrate had no effect. We conclude that PKCγ regulates ethanol-induced alterations in α4-containing GABA(A) receptors.
Collapse
Affiliation(s)
- David F. Werner
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - Sandeep Kumar
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - Hugh E. Criswell
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - Asha Suryanarayanan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - J. Alex Fetzer
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - Chris E. Comerford
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178
| |
Collapse
|
39
|
Kia A, Ribeiro F, Nelson R, Gavrilovici C, Ferguson SSG, Poulter MO. Kindling alters neurosteroid-induced modulation of phasic and tonic GABAA receptor-mediated currents: role of phosphorylation. J Neurochem 2011; 116:1043-56. [DOI: 10.1111/j.1471-4159.2010.07156.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|