1
|
Borruto AM, Calpe-López C, Spanagel R, Bernardi RE. Conditional deletion of the AMPA-GluA1 and NMDA-GluN1 receptor subunit genes in midbrain D1 neurons does not alter cocaine reward in mice. Neuropharmacology 2024; 258:110081. [PMID: 39002853 DOI: 10.1016/j.neuropharm.2024.110081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Synaptic plasticity in the mesolimbic dopamine (DA) system contributes to the neural adaptations underlying addictive behaviors and relapse. However, the specific behavioral relevance of glutamatergic excitatory drive onto dopamine D1 receptor (D1R)-expressing neurons in mediating the reinforcing effect of cocaine remains unclear. Here, we investigated how midbrain AMPAR and NMDAR function modulate cocaine reward-related behavior using mutant mouse lines lacking the glutamate receptor genes Gria1 or Grin1 in D1R-expressing neurons (GluA1D1CreERT2 or GluN1D1CreERT2, respectively). We found that conditional genetic deletion of either GluA1 or GluN1 within this neuronal sub-population did not impact the ability of acute cocaine injection to increase intracranial self-stimulation (ICSS) ratio or reduced brain reward threshold compared to littermate controls. Additionally, our data demonstrate that deletion of GluA1 and GluN1 receptor subunits within D1R-expressing neurons did not affect cocaine reinforcement in an operant self-administration paradigm, as mutant mice showed comparable cocaine responses and intake to controls. Given the pivotal role of glutamate receptors in mediating relapse behavior, we further explored the impact of genetic deletion of AMPAR and NMDAR onto D1R-expressing neurons on cue-induced reinstatement following extinction. Surprisingly, deletion of AMPAR and NMDAR onto these neurons did not impair cue-induced reinstatement of cocaine-seeking behavior. These findings suggest that glutamatergic activity via NMDAR and AMPAR in D1R-expressing neurons may not exclusively mediate the reinforcing effects of cocaine and cue-induced reinstatement.
Collapse
MESH Headings
- Animals
- Cocaine/pharmacology
- Cocaine/administration & dosage
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Reward
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Mice
- Self Administration
- Male
- Mesencephalon/metabolism
- Mesencephalon/drug effects
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Neurons/metabolism
- Neurons/drug effects
- Mice, Knockout
- Dopamine Uptake Inhibitors/pharmacology
- Mice, Inbred C57BL
- Reinforcement, Psychology
- Nerve Tissue Proteins
Collapse
Affiliation(s)
- Anna Maria Borruto
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Claudia Calpe-López
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; German Center for Mental Health (DZPG), Partner Site Mannheim, Heidelberg, Ulm, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
2
|
Oka M, Yoshino R, Kitanaka N, Hall FS, Uhl GR, Kitanaka J. Role of glycogen synthase kinase-3β in dependence and abuse liability of alcohol. Alcohol Alcohol 2024; 59:agad086. [PMID: 38145944 DOI: 10.1093/alcalc/agad086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Alcohol is a major abused drug worldwide that contributes substantially to health and social problems. These problems result from acute alcohol overuse as well as chronic use, leading to alcohol use disorder (AUD). A major goal of this field is to establish a treatment for alcohol abuse and dependence in patients with AUD. The central molecular mechanisms of acute alcohol actions have been extensively investigated in rodent models. AIMS One of the central mechanisms that may be involved is glycogen synthase kinase-3β (GSK-3β) activity, a key enzyme involved in glycogen metabolism but which has crucial roles in numerous cellular processes. Although the exact mechanisms leading from acute alcohol actions to these chronic changes in GSK-3β function are not yet clear, GSK-3β nonetheless constitutes a potential therapeutic target for AUD by reducing its function using GSK-3β inhibitors. This review is focused on the correlation between GSK-3β activity and the degree of alcohol consumption. METHODS Research articles regarding investigation of effect of GSK-3β on alcohol consumption in rodents were searched on PubMed, Embase, and Scopus databases using keywords "glycogen synthase kinase," "alcohol (or ethanol)," "intake (or consumption)," and evaluated by changes in ratios of pGSK-3βSer9/pGSK-3β. RESULTS In animal experiments, GSK-3β activity decreases in the brain under forced and voluntary alcohol consumption while GSK-3β activity increases under alcohol-seeking behavior. CONCLUSIONS Several pieces of evidence suggest that alterations in GSK-3β function are important mediators of chronic ethanol actions, including those related to alcohol dependence and the adverse effects of chronic ethanol exposure.
Collapse
Affiliation(s)
- Masahiro Oka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| | - Rui Yoshino
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| | - Nobue Kitanaka
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo 663-8501, Japan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Frederic and Mary Wolfe Center HEB 282D, Mail Stop 1015, 3000 Arlington Avenue,Toledo, OH 43614, United States
| | - George R Uhl
- Neurology Service, VA Maryland Healthcare System, 10 North Greene Street, Baltimore, MD 21201, United States
- Departments of Neurology and Pharmacology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junichi Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| |
Collapse
|
3
|
Pagano R, Salamian A, Zielinski J, Beroun A, Nalberczak-Skóra M, Skonieczna E, Cały A, Tay N, Banaschewski T, Desrivières S, Grigis A, Garavan H, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Poustka L, Hohmann S, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Kalita K, Bito H, Müller CP, Schumann G, Okuno H, Radwanska K. Arc controls alcohol cue relapse by a central amygdala mechanism. Mol Psychiatry 2023; 28:733-745. [PMID: 36357670 DOI: 10.1038/s41380-022-01849-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Alcohol use disorder (AUD) is a chronic and fatal disease. The main impediment of the AUD therapy is a high probability of relapse to alcohol abuse even after prolonged abstinence. The molecular mechanisms of cue-induced relapse are not well established, despite the fact that they may offer new targets for the treatment of AUD. Using a comprehensive animal model of AUD, virally-mediated and amygdala-targeted genetic manipulations by CRISPR/Cas9 technology and ex vivo electrophysiology, we identify a mechanism that selectively controls cue-induced alcohol relapse and AUD symptom severity. This mechanism is based on activity-regulated cytoskeleton-associated protein (Arc)/ARG3.1-dependent plasticity of the amygdala synapses. In humans, we identified single nucleotide polymorphisms in the ARC gene and their methylation predicting not only amygdala size, but also frequency of alcohol use, even at the onset of regular consumption. Targeting Arc during alcohol cue exposure may thus be a selective new mechanism for relapse prevention.
Collapse
Affiliation(s)
- Roberto Pagano
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ahmad Salamian
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Janusz Zielinski
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Anna Beroun
- Laboratory of Neuronal Plasticity, BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Maria Nalberczak-Skóra
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Edyta Skonieczna
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Cały
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Nicole Tay
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Rüdiger Brühl
- Braunschweig and Berlin, Physikalisch-Technische Bundesanstalt (PTB), Berlin, Germany
| | - Jean-Luc Martinot
- INSERM U1299 "Trajectoires développementales en psychiatrie, Institut National de la Santé et de la Recherche Médicale, Paris, Gif-sur-Yvette, France
- AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
- Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Université Paris-Saclay, Paris, Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- INSERM U1299 "Trajectoires développementales en psychiatrie, Institut National de la Santé et de la Recherche Médicale, Paris, Gif-sur-Yvette, France
- AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
- Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Université Paris-Saclay, Paris, Gif-sur-Yvette, France
| | - Eric Artiges
- INSERM U1299 "Trajectoires développementales en psychiatrie, Institut National de la Santé et de la Recherche Médicale, Paris, Gif-sur-Yvette, France
- AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
- Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Université Paris-Saclay, Paris, Gif-sur-Yvette, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | | | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Christian P Müller
- Section of Addiction Medicine, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Hiroyuki Okuno
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
4
|
Ray SK, Mukherjee S. Neuropharmacology of Alcohol Addiction with Special Emphasis on Proteomic Approaches for Identification of Novel Therapeutic Targets. Curr Neuropharmacol 2023; 21:119-132. [PMID: 35959616 PMCID: PMC10193758 DOI: 10.2174/1570159x20666220811092906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/01/2022] [Accepted: 07/10/2022] [Indexed: 02/04/2023] Open
Abstract
Alcohol is a generic pharmacological agent with only a few recognized primary targets. Nmethyl- D-aspartate, gamma-aminobutyric acid, glycine, 5-hydroxytryptamine 3 (serotonin), nicotinic acetylcholine receptors, and L-type Ca2+ channels and G-protein-activated inwardly rectifying K channels are all involved. Following the first hit of alcohol on specific brain targets, the second wave of indirect effects on various neurotransmitter/neuropeptide systems begins, leading to the typical acute behavioral effects of alcohol, which range from disinhibition to sedation and even hypnosis as alcohol concentrations rise. Recent research has revealed that gene regulation is significantly more complex than previously thought and does not fully explain changes in protein levels. As a result, studying the proteome directly, which differs from the genome/transcriptome in terms of complexity and dynamicity, has provided unique insights into extraordinary advances in proteomic techniques that have changed the way we can analyze the composition, regulation, and function of protein complexes and pathways underlying altered neurobiological conditions. Neuroproteomics has the potential to revolutionize alcohol research by allowing researchers to gain a better knowledge of how alcohol impacts protein structure, function, connections, and networks on a global scale. The amount of information collected from these breakthroughs can aid in identifying valuable biomarkers for early detection and improved prognosis of an alcohol use disorder and future pharmaceutical targets for the treatment of alcoholism.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Science, Bhopal, Madhya Pradesh 462020, India
| |
Collapse
|
5
|
Wu CT, Chaffin AT, Ryan KK. Fibroblast Growth Factor 21 Facilitates the Homeostatic Control of Feeding Behavior. J Clin Med 2022; 11:580. [PMID: 35160033 PMCID: PMC8836936 DOI: 10.3390/jcm11030580] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a stress hormone that is released from the liver in response to nutritional and metabolic challenges. In addition to its well-described effects on systemic metabolism, a growing body of literature now supports the notion that FGF21 also acts via the central nervous system to control feeding behavior. Here we review the current understanding of FGF21 as a hormone regulating feeding behavior in rodents, non-human primates, and humans. First, we examine the nutritional contexts that induce FGF21 secretion. Initial reports describing FGF21 as a 'starvation hormone' have now been further refined. FGF21 is now better understood as an endocrine mediator of the intracellular stress response to various nutritional manipulations, including excess sugars and alcohol, caloric deficits, a ketogenic diet, and amino acid restriction. We discuss FGF21's effects on energy intake and macronutrient choice, together with our current understanding of the underlying neural mechanisms. We argue that the behavioral effects of FGF21 function primarily to maintain systemic macronutrient homeostasis, and in particular to maintain an adequate supply of protein and amino acids for use by the cells.
Collapse
Affiliation(s)
| | | | - Karen K. Ryan
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA 95616, USA; (C.-T.W.); (A.T.C.)
| |
Collapse
|
6
|
Bowen MT, George O, Muskiewicz DE, Hall FS. FACTORS CONTRIBUTING TO THE ESCALATION OF ALCOHOL CONSUMPTION. Neurosci Biobehav Rev 2022; 132:730-756. [PMID: 34839930 PMCID: PMC8892842 DOI: 10.1016/j.neubiorev.2021.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 01/03/2023]
Abstract
Understanding factors that contribute to the escalation of alcohol consumption is key to understanding how an individual transitions from non/social drinking to AUD and to providing better treatment. In this review, we discuss how the way ethanol is consumed as well as individual and environmental factors contribute to the escalation of ethanol consumption from intermittent low levels to consistently high levels. Moreover, we discuss how these factors are modelled in animals. It is clear a vast array of complex, interacting factors influence changes in alcohol consumption. Some of these factors act early in the acquisition of ethanol consumption and initial escalation, while others contribute to escalation of ethanol consumption at a later stage and are involved in the development of alcohol dependence. There is considerable need for more studies examining escalation associated with the formation of dependence and other hallmark features of AUD, especially studies examining mechanisms, as it is of considerable relevance to understanding and treating AUD.
Collapse
Affiliation(s)
- Michael T. Bowen
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, 2050, Australia,The University of Sydney, Faculty of Science, School of Psychology, Sydney, NSW, 2006, Australia,Corresponding Author: Michael T. Bowen, Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, Sydney, NSW, 2050, Australia,
| | - Olivier George
- Department of Psychology, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Dawn E. Muskiewicz
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacology and Pharmacological Science, University of Toledo, OH, USA
| | - F. Scott Hall
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacology and Pharmacological Science, University of Toledo, OH, USA
| |
Collapse
|
7
|
Stafford AM, Yamamoto BK, Phillips TJ. Combined and sequential effects of alcohol and methamphetamine in animal models. Neurosci Biobehav Rev 2021; 131:248-269. [PMID: 34543650 PMCID: PMC8642292 DOI: 10.1016/j.neubiorev.2021.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 11/16/2022]
Abstract
Comorbid drug use, often alcohol with other drugs, poses significant health and societal concerns. Methamphetamine is among the illicit drugs most often co-used with alcohol. The current review examines the animal literature for impacts of comorbid alcohol and methamphetamine exposure. We found evidence for additive or synergistic effects of combined or sequential exposure on behavior and physiology. Dopaminergic, serotonergic, and glutamatergic systems are all impacted by combined exposure to alcohol and methamphetamine and cyclooxygenase-2 activity plays an important role in their combined neurotoxic effects. Adverse consequences of comorbid exposure include altered brain development with prenatal exposure, impaired learning and memory, motor deficits, gastrotoxicity, hepatotoxicity, and augmented intake under some conditions. Given high susceptibility to drug experimentation in adolescence, studies of co-exposure during the adolescent period and of how adolescent exposure to one drug impacts later use or sensitivity to the other drug should be a priority. Further, to gain traction on prevention and treatment, additional research to identify motivational and neurobiological drivers and consequences of comorbid use is needed.
Collapse
Affiliation(s)
- Alexandra M Stafford
- Department of Behavioral Neuroscience, Portland Alcohol Abuse Research Center and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA.
| | - Bryan K Yamamoto
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience, Portland Alcohol Abuse Research Center and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA; Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
8
|
Faccidomo S, Cogan ES, Hon OJ, Hoffman JL, Saunders BL, Eastman VR, Kim M, Taylor SM, McElligott ZA, Hodge CW. Calcium-permeable AMPA receptor activity and GluA1 trafficking in the basolateral amygdala regulate operant alcohol self-administration. Addict Biol 2021; 26:e13049. [PMID: 33955100 DOI: 10.1111/adb.13049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/16/2021] [Indexed: 12/22/2022]
Abstract
Addiction is viewed as maladaptive glutamate-mediated neuroplasticity that is regulated, in part, by calcium-permeable AMPA receptor (CP-AMPAR) activity. However, the contribution of CP-AMPARs to alcohol-seeking behavior remains to be elucidated. We evaluated CP-AMPAR activity in the basolateral amygdala (BLA) as a potential target of alcohol that also regulates alcohol self-administration in C57BL/6J mice. Operant self-administration of sweetened alcohol increased spontaneous EPSC frequency in BLA neurons that project to the nucleus accumbens as compared with behavior-matched sucrose controls indicating an alcohol-specific upregulation of synaptic activity. Bath application of the CP-AMPAR antagonist NASPM decreased evoked EPSC amplitude only in alcohol self-administering mice indicating alcohol-induced synaptic insertion of CP-AMPARs in BLA projection neurons. Moreover, NASPM infusion in the BLA dose-dependently decreased the rate of operant alcohol self-administration providing direct evidence for CP-AMPAR regulation of alcohol reinforcement. As most CP-AMPARs are GluA1-containing, we asked if alcohol alters the activation state of GluA1-containing AMPARs. Immunocytochemistry results showed elevated GluA1-S831 phosphorylation in the BLA of alcohol as compared with sucrose mice. To investigate mechanistic regulation of alcohol self-administration by GluA1-containing AMPARs, we evaluated the necessity of GluA1 trafficking using a TET-ON AAV encoding a dominant-negative GluA1 c-terminus (GluA1ct) that blocks activity-dependent synaptic delivery of native GluA1-containing AMPARs. GluA1ct expression in the BLA reduced alcohol self-administration with no effect on sucrose controls. These results show that CP-AMPAR activity and GluA1 trafficking in the BLA mechanistically regulate the reinforcing effects of sweetened alcohol. Pharmacotherapeutic targeting these mechanisms of maladaptive neuroplasticity may aid medical management of alcohol use disorder.
Collapse
Affiliation(s)
- Sara Faccidomo
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Elizabeth S. Cogan
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Olivia J. Hon
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Neuroscience Curriculum The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Jessica L. Hoffman
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Briana L. Saunders
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Vallari R. Eastman
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Michelle Kim
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Seth M. Taylor
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Clyde W. Hodge
- Bowles Center for Alcohol Studies The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
9
|
Ketamine Modulates the Neural Correlates of Reward Processing in Unmedicated Patients in Remission from Depression. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:285-292. [PMID: 34126264 DOI: 10.1016/j.bpsc.2021.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/26/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Ketamine as an antidepressant improves anhedonia as early as 2h post-infusion. These drug effects are thought to be exerted via actions on reward-related brain areas-yet, these actions remain largely unknown. Our study investigates ketamine's effects during the anticipation and receipt of an expected reward, after the psychotomimetic effects of ketamine have passed, when early antidepressant effects are reported. METHODS We examined ketamine's effects during the anticipation and receipt of expected rewards on pre-defined brain areas, namely the dorsal and ventral striatum, the ventral tegmental area, the amygdala and the insula. We have recruited 37 male and female participants who remitted from depression and were free from symptoms and antidepressant treatments at the time of the scan. Participants were scanned, 2h after drug administration, in a double-blind cross over design (ketamine:0.5mg/kg and placebo) while performing a monetary reward task. RESULTS A significant main effect of ketamine, across all ROIs, was observed during the anticipation and feedback phases of win and no-win trials. The drug effects were particularly prominent in the nucleus accumbens and putamen, which showed increased activation upon the receipt of smaller rewards compared to neutral. The levels of (2R,6R)-HNK, 2h post-infusion, significantly correlated with the activation observed in the ventral tegmental area for that contrast. CONCLUSIONS These findings demonstrate that ketamine can produce detectable changes in reward-related brain areas, 2h after infusion, which occur without symptom changes and support the idea that ketamine might improve reward-related symptoms via modulation of response to feedback.
Collapse
|
10
|
NMDA Receptors in Accumbal D1 Neurons Influence Chronic Sugar Consumption and Relapse. eNeuro 2021; 8:ENEURO.0029-21.2021. [PMID: 33906970 PMCID: PMC8143023 DOI: 10.1523/eneuro.0029-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 11/29/2022] Open
Abstract
Glutamatergic input via NMDA and AMPA receptors within the mesolimbic dopamine (DA) pathway plays a critical role in the development of addictive behavior and relapse toward drugs of abuse. Although well-established for drugs of abuse, it is not clear whether glutamate receptors within the mesolimbic system are involved in mediating chronic consumption and relapse following abstinence from a non-drug reward. Here, we evaluated the contribution of mesolimbic glutamate receptors in mediating chronic sugar consumption and the sugar-deprivation effect (SDE), which is used as a measure of relapse-like behavior following abstinence. We studied four inducible mutant mouse lines lacking the GluA1 or GluN1 subunit in either DA transporter (DAT) or D1R-expressing neurons in an automated monitoring system for free-choice sugar drinking in the home cage. Mice lacking either GluA1 or GluN1 in D1R-expressing neurons (GluA1D1CreERT2 or GluN1D1CreERT2mice) have altered sugar consumption in both sexes, whereas GluA1DATCreERT2 and GluN1DATCreERT2do not differ from their respective littermate controls. In terms of relapse-like behavior, female GluN1D1CreERT2mice show a more pronounced SDE. Given that glutamate receptors within the mesolimbic system play a critical role in mediating relapse behavior of alcohol and other drugs of abuse, it is surprising that these receptors do not mediate the SDE, or in the case of female GluN1D1CreERT2 mice, show an opposing effect. We conclude that a relapse-like phenotype of sugar consumption differs from that of drugs of abuse on the molecular level, at least with respect to the contribution of mesolimbic glutamate receptors.
Collapse
|
11
|
Domi E, Domi A, Adermark L, Heilig M, Augier E. Neurobiology of alcohol seeking behavior. J Neurochem 2021; 157:1585-1614. [PMID: 33704789 DOI: 10.1111/jnc.15343] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/29/2022]
Abstract
Alcohol addiction is a chronic relapsing brain disease characterized by an impaired ability to stop or control alcohol use despite adverse consequences. A main challenge of addiction treatment is to prevent relapse, which occurs in more than >50% of newly abstinent patients with alcohol disorder within 3 months. In people suffering from alcohol addiction, stressful events, drug-associated cues and contexts, or re-exposure to a small amount of alcohol trigger a chain of behaviors that frequently culminates in relapse. In this review, we first present the preclinical models that were developed for the study of alcohol seeking behavior, namely the reinstatement model of alcohol relapse and compulsive alcohol seeking under a chained schedule of reinforcement. We then provide an overview of the neurobiological findings obtained using these animal models, focusing on the role of opioids systems, corticotropin-release hormone and neurokinins, followed by dopaminergic, glutamatergic, and GABAergic neurotransmissions in alcohol seeking behavior.
Collapse
Affiliation(s)
- Esi Domi
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Heilig
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Eric Augier
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| |
Collapse
|
12
|
Female mice are more prone to develop an addictive-like phenotype for sugar consumption. Sci Rep 2021; 11:7364. [PMID: 33795734 PMCID: PMC8016940 DOI: 10.1038/s41598-021-86797-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
The concept of “sugar addiction” is gaining increasing attention in both the lay media and scientific literature. However, the concept of sugar addiction is controversial and only a few studies to date have attempted to determine the “addictive” properties of sugar using rigorous scientific criteria. Here we set out to systematically test the addictive properties of sugar in male and female mice using established paradigms and models from the drug addiction field. Male and female C57BL/6N (8–10 weeks old) were evaluated in 4 experimental procedures to study the addictive properties of sugar: (i) a drinking in the dark (DID) procedure to model sugar binging; (ii) a long-term free choice home cage drinking procedure measuring the sugar deprivation effect (SDE) following an abstinence phase; (iii) a long-term operant sugar self-administration with persistence, motivation and compulsivity measures and (iv) intracranial self-stimulation (ICSS). Female mice were more vulnerable to the addictive properties of sugar than male mice, showing higher binge and long-term, excessive drinking, a more pronounced relapse-like drinking following deprivation, and higher persistence and motivation for sugar. No sex differences were seen in a compulsivity test or reward sensitivity measured using ICSS following extended sugar consumption. This study demonstrates the occurrence of an addictive-like phenotype for sugar in male and female mice, similar to drugs of abuse, and suggests sex-dependent differences in the development of sugar addiction.
Collapse
|
13
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
14
|
Alasmari F, Alhaddad H, Wong W, Bell RL, Sari Y. Ampicillin/Sulbactam Treatment Modulates NMDA Receptor NR2B Subunit and Attenuates Neuroinflammation and Alcohol Intake in Male High Alcohol Drinking Rats. Biomolecules 2020; 10:biom10071030. [PMID: 32664441 PMCID: PMC7407831 DOI: 10.3390/biom10071030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
Exposure to ethanol commonly manifests neuroinflammation. Beta (β)-lactam antibiotics attenuate ethanol drinking through upregulation of astroglial glutamate transporters, especially glutamate transporter-1 (GLT-1), in the mesocorticolimbic brain regions, including the nucleus accumbens (Acb). However, the effect of β-lactam antibiotics on neuroinflammation in animals chronically exposed to ethanol has not been fully investigated. In this study, we evaluated the effects of ampicillin/sulbactam (AMP/SUL, 100 and 200 mg/kg, i.p.) on ethanol consumption in high alcohol drinking (HAD1) rats. Additionally, we investigated the effects of AMP/SUL on GLT-1 and N-methyl-d-aspartate (NMDA) receptor subtypes (NR2A and NR2B) in the Acb core (AcbCo) and Acb shell (AcbSh). We found that AMP/SUL at both doses attenuated ethanol consumption and restored ethanol-decreased GLT-1 and NR2B expression in the AcbSh and AcbCo, respectively. Moreover, AMP/SUL (200 mg/kg, i.p.) reduced ethanol-increased high mobility group box 1 (HMGB1) and receptor for advanced glycation end-products (RAGE) expression in the AcbSh. Moreover, both doses of AMP/SUL attenuated ethanol-elevated tumor necrosis factor-alpha (TNF-α) in the AcbSh. Our results suggest that AMP/SUL attenuates ethanol drinking and modulates NMDA receptor NR2B subunits and HMGB1-associated pathways.
Collapse
Affiliation(s)
- Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Woonyen Wong
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
| | - Richard L. Bell
- Department of Psychiatry and Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: (R.L.B.); (Y.S.); Tel.: +317-278-8407 (R.L.B.); +419-383-1507 (Y.S.)
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (H.A.); (W.W.)
- Correspondence: (R.L.B.); (Y.S.); Tel.: +317-278-8407 (R.L.B.); +419-383-1507 (Y.S.)
| |
Collapse
|
15
|
Zhou Y, Liang Y, Kreek MJ. mTORC1 pathway is involved in the kappa opioid receptor activation-induced increase in excessive alcohol drinking in mice. Pharmacol Biochem Behav 2020; 195:172954. [PMID: 32470351 DOI: 10.1016/j.pbb.2020.172954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
KOP-r agonist U50,488H produces strong aversion and anxiety/depression-like behaviors that enhance alcohol intake and promote alcohol seeking and relapse-like drinking in rodents. Mammalian target of rapamycin complex 1 (mTORC1) pathway in mouse striatum is highly involved in excessive alcohol intake and seeking, and in the U50,488H-induced conditioned place aversion. Therefore, we hypothesized that KOP-r activation increases alcohol consumption through the mTORC1 activation. This study focuses on: (1) how chronic excessive alcohol drinking (4-day drinking-in-the-dark paradigm followed by 3-week chronic intermittent access drinking paradigm [two-bottle choice, 24-h access every other day]) affected nuclear transcript levels of the mTORC1 pathway genes in mouse nucleus accumbens shell (NAcs), using transcriptome-wide RNA sequencing analysis; and (2) whether selective mTORC1 inhibitor rapamycin could alter excessive alcohol drinking and prevent U50,488H-promoted alcohol intake. Thirteen nuclear transcripts of mTORC1 pathway genes showed significant up-regulation in the NAcs, with two genes down-regulated, after excessive alcohol drinking, suggesting the mTORC1 pathway was profoundly disrupted. Single administration of rapamycin decreased alcohol drinking in a dose-dependent manner. U50,488H increased alcohol drinking, and pretreatment with rapamycin, at a dose lower than effective doses, blocked the U50,488H-promoted alcohol intake in a dose-dependent manner, indicating a mTORC1-mediated mechanism. Our results provide supportive and direct evidence relevant to the transcriptional profiling of the critical mTORC1 genes in mouse NAc shell: with functional and pharmacological effects of rapamycin, altered nuclear transcripts in the mTORC1 signaling pathway after excessive alcohol drinking may contribute to increased alcohol intake triggered by KOP-r activation.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, USA.
| | - Yupu Liang
- Research Bioinformatics, CCTS, The Rockefeller University, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, USA
| |
Collapse
|
16
|
Wei S, Li Y, Gong Q, Liang H, Liu Q, Bernardi RE, Zhang H, Chen F, Lawrence AJ, Liang J. Brucine N‐Oxide Reduces Ethanol Intake and Preference in Alcohol‐Preferring Male Fawn‐Hooded Rats. Alcohol Clin Exp Res 2020; 44:1321-1328. [PMID: 32343845 DOI: 10.1111/acer.14344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/18/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Shoupeng Wei
- From the Department of Pharmacology (SW, QG, HL, QL) School of Basic Medicine Sciences Peking University Beijing China
| | - Yu‐ling Li
- Department of Pharmacy (Y‐LL) East Hospital Tongji University School of Medicine Shanghai China
| | - Qi Gong
- From the Department of Pharmacology (SW, QG, HL, QL) School of Basic Medicine Sciences Peking University Beijing China
| | - Hui Liang
- From the Department of Pharmacology (SW, QG, HL, QL) School of Basic Medicine Sciences Peking University Beijing China
| | - Qing Liu
- From the Department of Pharmacology (SW, QG, HL, QL) School of Basic Medicine Sciences Peking University Beijing China
| | - Rick E. Bernardi
- Institute of Psychopharmacology (REB) Central Institute of Mental Health Medical Faculty Mannheim University of Heidelberg Mannheim Germany
| | - Han‐Ting Zhang
- Departments of Behavioral Medicine & Psychiatry and Physiology & Pharmacology (H‐TZ) West Virginia University Health Sciences Center Morgantown West Virginia
| | - Feng Chen
- The Florey Institute of Neuroscience and Mental Health (FC, AJL) University of Melbourne Parkville3010Victoria Australia
| | - Andrew J. Lawrence
- The Florey Institute of Neuroscience and Mental Health (FC, AJL) University of Melbourne Parkville3010Victoria Australia
| | - Jian‐hui Liang
- Department of Molecular and Cellular Pharmacology (J‐hL) School of Pharmaceutical Sciences Peking University Beijing China
| |
Collapse
|
17
|
Bilbao A, Neuhofer D, Sepers M, Wei SP, Eisenhardt M, Hertle S, Lassalle O, Ramos-Uriarte A, Puente N, Lerner R, Thomazeau A, Grandes P, Lutz B, Manzoni OJ, Spanagel R. Endocannabinoid LTD in Accumbal D1 Neurons Mediates Reward-Seeking Behavior. iScience 2020; 23:100951. [PMID: 32179475 PMCID: PMC7068121 DOI: 10.1016/j.isci.2020.100951] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/15/2019] [Accepted: 02/24/2020] [Indexed: 11/30/2022] Open
Abstract
The nucleus accumbens (NAc) plays a key role in drug-related behavior and natural reward learning. Synaptic plasticity in dopamine D1 and D2 receptor medium spiny neurons (MSNs) of the NAc and the endogenous cannabinoid (eCB) system have been implicated in reward seeking. However, the precise molecular and physiological basis of reward-seeking behavior remains unknown. We found that the specific deletion of metabotropic glutamate receptor 5 (mGluR5) in D1-expressing MSNs (D1miRmGluR5 mice) abolishes eCB-mediated long-term depression (LTD) and prevents the expression of drug (cocaine and ethanol), natural reward (saccharin), and brain-stimulation-seeking behavior. In vivo enhancement of 2-arachidonoylglycerol (2-AG) eCB signaling within the NAc core restores both eCB-LTD and reward-seeking behavior in D1miRmGluR5 mice. The data suggest a model where the eCB and glutamatergic systems of the NAc act in concert to mediate reward-seeking responses. mGluR5-D1-CB1-induced eCB-LTD mediates drugs of abuse and natural reward seeking eCB-LTD in D2-MSNs plays no important role in processing of reward-seeking responses Loss of eCB-LTD is a consequence of higher MAGL activity and lower CB1R expression Acute drug administration stops craving for alternative rewards on following days
Collapse
Affiliation(s)
- Ainhoa Bilbao
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany.
| | - Daniela Neuhofer
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Marja Sepers
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Shou-Peng Wei
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Manuela Eisenhardt
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Sarah Hertle
- Behavioral Genetics Research Group, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Olivier Lassalle
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, 107 S Indiana Avenue, Bloomington, IN 47405, USA
| | - Almudena Ramos-Uriarte
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Raissa Lerner
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Aurore Thomazeau
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Olivier J Manzoni
- INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273, Marseille Cedex 09, France; Aix-Marseille University, Jardindu Pharo, 58 Boulevard Charles Livon, Marseille, 13007, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Indiana University, 107 S Indiana Avenue, Bloomington, IN 47405, USA.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Medical Faculty Mannheim, 68159 Mannheim, Germany.
| |
Collapse
|
18
|
Bilbao A, Leixner S, Wei S, Cantacorps L, Valverde O, Spanagel R. Reduced sensitivity to ethanol and excessive drinking in a mouse model of neuropathic pain. Addict Biol 2019; 24:1008-1018. [PMID: 31237390 DOI: 10.1111/adb.12784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The co-occurrence of chronic pain and alcohol use disorders (AUDs) involves complex interactions between genetic and neurophysiological aspects, and the research has reported mixed findings when they both co-occur. There is also an indication of a gender-dependent effect; males are more likely to use alcohol to cope with chronic pain problems than females. Recently, a new conceptualization has emerged, proposing that the negative affective component of pain drives and maintains alcohol-related behaviors. We studied in a longitudinal fashion alterations in alcohol drinking patterns and pain thresholds in a mouse model of chronic neuropathic pain in a sex-dependent manner. Following partial denervation (spared nerve injury [SNI]), stimulus-evoked pain responses were measured before chronic alcohol consumption, during drinking, during a deprivation phase, and following an episode of excessive drinking. During the course of alcohol drinking, we observed pronounced sex differences in pain thresholds. Male mice showed a strong increase in pain thresholds, suggesting an analgesic effect induced by alcohol over time, an effect that was not observed in female mice. SNI mice did not differ from sham-operated controls in baseline alcohol consumption. However, following a deprivation phase and the reintroduction of ethanol, male SNI mice but not female mice showed more pronounced excessive drinking than controls. Finally, we observed decreased central ethanol sensitivity in male SNI mice but not in females. Together with our finding, that ethanol is able to decrease a pain-induced negative affective memory we come to following conclusion. We propose that a lower sensitivity to the intoxicating effects of alcohol together with the ability of alcohol to reduce the negative affective component of pain may explain the higher co-occurrence of AUD in male chronic pain patients.
Collapse
Affiliation(s)
- Ainhoa Bilbao
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim; Heidelberg University; Mannheim Germany
| | - Sarah Leixner
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim; Heidelberg University; Mannheim Germany
| | - Shoupeng Wei
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim; Heidelberg University; Mannheim Germany
| | - Lídia Cantacorps
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim; Heidelberg University; Mannheim Germany
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, IMIM (Hospital del Mar Medical Research Institute); Pompeu Fabra University; Barcelona Spain
| | - Olga Valverde
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, IMIM (Hospital del Mar Medical Research Institute); Pompeu Fabra University; Barcelona Spain
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim; Heidelberg University; Mannheim Germany
| |
Collapse
|
19
|
Shi Z, Xie Y, Ren H, He B, Wang M, Wan J, Yuan T, Yao X, Su H. Fish oil treatment reduces chronic alcohol exposure induced synaptic changes. Addict Biol 2019; 24:577-589. [PMID: 29569345 DOI: 10.1111/adb.12623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/22/2018] [Accepted: 02/27/2018] [Indexed: 12/16/2022]
Abstract
Alcohol addiction is a chronic neuropsychiatric disorder that represents one of the most serious global public health problems. Yet, currently there still lacks an effective pharmacotherapy. Omega-3 polyunsaturated fatty acids (N-3 PUFAs) have exhibited beneficial effects in a variety of neurological disorders, particularly in reversing behavioral deficits and neurotoxicity induced by prenatal alcohol exposure and binge drinking. In the present study, we investigated if fish oil, which is rich in N-3 PUFAs, had beneficial effects on preventing relapse and alleviating withdrawal symptoms after chronic alcohol exposure. Our results demonstrated that fish oil significantly reduced the chronic alcohol exposure-induced aberrant dendritic morphologic changes of the medium-sized spiny neurons in the core and the shell of nucleus accumbens. This inhibited the expression of AMPAR2-lacking AMPARs and their accumulation on the post synaptic membranes of medium-sized spiny neurons and eventually alleviated withdrawal symptoms and alcohol dependence. Our study therefore suggests that N-3 PUFAs are promising for treating withdrawal symptoms and alcohol dependence.
Collapse
Affiliation(s)
- Zhe Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of Macau China
| | - Youna Xie
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated HospitalSun Yat‐Sen University China
| | - Huixia Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of Macau China
| | - Baixuan He
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated HospitalSun Yat‐Sen University China
| | - Meng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of Macau China
| | - Jian‐Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of Macau China
| | - Ti‐Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of Medicine China
- Co‐innovation Center of NeuroregenerationNantong University China
| | - Xiaoli Yao
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated HospitalSun Yat‐Sen University China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of Macau China
| |
Collapse
|
20
|
Lainiola M, Hietala L, Linden AM, Aitta-Aho T. The lack of conditioned place preference, but unaltered stimulatory and ataxic effects of alcohol in mGluR3-KO mice. J Psychopharmacol 2019; 33:855-864. [PMID: 31070489 DOI: 10.1177/0269881119844178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Alcohol use associates with environmental cues that can later reinstate drinking patterns without any alcohol exposure. Alcohol-induced reward, when combined with contextual signals of various sensory modalities in the central synapses of mesolimbic reward circuitries, can lead to the formation of conditioned responses. AIMS As the activation of glutamatergic synapses is pivotal in such processes, we aimed to investigate whether the metabotropic glutamate receptor subtype 3 plays a role in alcohol-induced behaviours including place preference. METHODS The metabotropic glutamate receptor subtype 3 knockout (mGluR3-KO) mouse line was used to study alcohol-induced place preference, locomotor activating and ataxic effects, limited access alcohol drinking, and preference for sucrose and saccharin. RESULTS Alcohol-induced horizontal locomotor stimulation and reduced rearing behaviour remained unchanged in the mGluR3-KO mice. However, alcohol-induced place conditioning in an unbiased paradigm setup was lacking in the mGluR3-KO mice, but clearly present in wildtype mice. Locomotor activity was not different between the mGluR3-KO and wildtype mice during the acquisition and expression trials. Alcohol consumption, studied through the 'drinking in the dark' model, remained unchanged in the mGluR3-KO mice, although low consumption in both wildtype and knockout mice hampers interpretation. The mGluR3-KO mice also showed normal sucrose and saccharin preference. CONCLUSIONS These studies indicate a role for metabotropic glutamate receptor subtype 3 in the conditioned contextual alcohol responses, but not in stimulatory, and ataxic alcohol effects.
Collapse
Affiliation(s)
- Mira Lainiola
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Lana Hietala
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anni-Maija Linden
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
21
|
Wegner SA, Hu B, De Oliveira Sergio T, Darevsky D, Kwok CCY, Lei K, Hopf FW. A novel NMDA receptor-based intervention to suppress compulsion-like alcohol drinking. Neuropharmacology 2019; 157:107681. [PMID: 31251994 DOI: 10.1016/j.neuropharm.2019.107681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 01/09/2023]
Abstract
Compulsive drives for alcohol, where intake persists despite adverse consequences, are substantial obstacles to treating Alcohol Use Disorder (AUD). However, there are limited treatment options and thus considerable interest in identifying new, potent and safe pharmacotherapies. We found that non-canonical N-methyl-d-aspartate receptors (NMDARs), active at hyperpolarized potentials, drive compulsion-like alcohol drinking in rats without affecting regular, alcohol-only intake. Congruent human studies suggest that NMDAR inhibition reduces alcohol drinking in treatment-seekers but not non-treatment-seekers and suppresses craving. These cross-species studies of consumption under conflict indicate that inhibiting non-canonical NMDARs could be of clinical value for AUD. d-serine activates NMDARs overall, but actually inhibits non-canonical NMDARs. Also, d-serine has been widely tested in humans as a moderate NMDAR modulator, but some nephrotoxicity concerns remain, and thus any strategy that reduces d-serine exposure could be of broad utility. Here, co-administration of sodium benzoate (NaBenz), which reduces d-serine breakdown, allowed subthreshold d-serine levels to suppress compulsion-like alcohol drinking without altering normal alcohol-only consumption, providing a novel intervention for AUD and underscoring the importance of non-canonical NMDARs for compulsion-like intake. Low NaBenz doses alone had no average effect on intake. NaBenz/d-serine reduced compulsion-like intake in nearly all animals, while higher d-serine alone decreased compulsion-like intake with less of an effect in lower-drinking subjects. Thus, combining subthreshold NaBenz and d-serine suppressed compulsion-like intake, helping both to alleviate some d-serine concerns, and, importantly, to reduce consequence-resistant consumption across nearly all individuals. Therefore, NaBenz/d-serine likely represents an FDA-approved and immediately-accessible pharmacotherapy to help counteract compulsion-like drives and treat AUD.
Collapse
Affiliation(s)
- Scott Andrew Wegner
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Bing Hu
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Thatiane De Oliveira Sergio
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - David Darevsky
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Claudina Choi-Yan Kwok
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Kelly Lei
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Frederic Woodward Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA; Wheeler Center for the Study of Addiction, University of California at San Francisco, USA.
| |
Collapse
|
22
|
Ballesta A, Orio L, Arco R, Vargas A, Romero-Sanchiz P, Nogueira-Arjona R, de Heras RG, Antón M, Ramírez-López M, Serrano A, Pavón FJ, de Fonseca FR, Suárez J, Alen F. Bupropion, a possible antidepressant without negative effects on alcohol relapse. Eur Neuropsychopharmacol 2019; 29:756-765. [PMID: 31064683 DOI: 10.1016/j.euroneuro.2019.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 12/15/2022]
Abstract
RATIONALE the role that antidepressants play on alcohol consumption is not well understood. Previous studies have reported that treatment with a Selective Serotonin Reuptake Inhibitor (SSRIs) increases alcohol consumption in an animal model of relapse, however it is unknown whether this effect holds for other antidepressants such as the atypical dopamine/norepinephrine reuptake inhibitors (SNDRI). OBJECTIVES the main goal of the present study was to compare the effects of two classes of antidepressants drugs, bupropion (SNDRI) and fluoxetine (SSRI), on alcohol consumption during relapse. Since glutamatergic and endocannabinoid signaling systems plays an important role in alcohol abuse and relapse, we also evaluated the effects of both antidepressants onthe expression of the main important genes and proteins of both systems in the prefrontal cortex, a critical brain region in alcohol relapse. METHODS rats were trained to self-administered alcohol. During abstinence, rats received a 14d-treatment with vehicle, fluoxetine (10 mg/kg) or bupropion (20 mg/kg), and we evaluated alcohol consumption during relapse for 3 weeks. Samples of prefrontal cortex were taken to evaluate the mRNA and protein expression of the different components of glutamatergic and endocannabinoid signaling systems. RESULTS fluoxetine treatment induced a long-lasting increase in alcohol consumption during relapse, an effect that was not observed in the case of bupropion treatment. The observed increases in alcohol consumption were accompanied by distinct alterations in the glutamate and endocannabinoid systems. CONCLUSIONS our results suggest that SSRIs can negatively impact alcohol consumption in relapse while SNDRIs have no effects. The observed increase in alcohol consumption are accompanied by functional alterations in the glutamatergic and endocannabinoid systems. This finding could open new strategies for the treatment of depression in patients with alcohol use disorders.
Collapse
Affiliation(s)
- Antonio Ballesta
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain
| | - Laura Orio
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain
| | - Rocío Arco
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain
| | - Antonio Vargas
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain
| | - Pablo Romero-Sanchiz
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain; Unidad de Salud Mental, Hospital Universitario Regional de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Spain; Departamento de Personalidad, Evaluación y Tratamientos Psicológicos. Universidad de Málaga, Málaga, Spain
| | - Raquel Nogueira-Arjona
- Unidad de Salud Mental, Hospital Universitario Regional de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Spain; Department of Psychology and Neuroscience, Dalhousie University, Canada
| | - Raquel Gómez de Heras
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain
| | - María Antón
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain
| | - Mayte Ramírez-López
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain
| | - Antonia Serrano
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain
| | - Francisco Javier Pavón
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain
| | - Fernando Rodríguez de Fonseca
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain; Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain.
| | - Juan Suárez
- Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain.
| | - Francisco Alen
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28224 Spain; Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental, Hospital Regional Universitario de Málaga, Av. Carlos Haya 82, sótano, Málaga 29010, Spain.
| |
Collapse
|
23
|
Lei K, Kwok C, Darevsky D, Wegner SA, Yu J, Nakayama L, Pedrozo V, Anderson L, Ghotra S, Fouad M, Hopf FW. Nucleus Accumbens Shell Orexin-1 Receptors Are Critical Mediators of Binge Intake in Excessive-Drinking Individuals. Front Neurosci 2019; 13:88. [PMID: 30814925 PMCID: PMC6381036 DOI: 10.3389/fnins.2019.00088] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/25/2019] [Indexed: 12/29/2022] Open
Abstract
Excessive, binge alcohol drinking is a potent and pernicious obstacle to treating alcohol use disorder (AUD), and heavy-drinking humans are responsible for much of the substantial costs and harms of AUD. Thus, identifying key mechanisms that drive intake in higher-drinking individuals may provide important, translationally useful therapeutic interventions. Orexin-1-receptors (Ox1Rs) promote states of high motivation, and studies with systemic Ox1R inhibition suggest a particular role in individuals with higher intake levels. However, little has been known about circuits where Ox1Rs promote pathological intake, especially excessive alcohol consumption. We previously discovered that binge alcohol drinking requires Ox1Rs in medial nucleus accumbens shell (Shell), using two-bottle-choice Drinking-in-the-Dark (2bc-DID) in adult, male C57BL/6 mice. Here, we show that Shell Ox1Rs promoted intake during intermittent-access alcohol drinking as well as 2bc-DID, and that Shell inhibition with muscimol/baclofen also suppressed 2bc-DID intake. Importantly, with this large data set, we were able to demonstrate that Shell Ox1Rs and overall activity were particularly important for driving alcohol consumption in higher-drinking individuals, with little overall impact in moderate drinkers. Shell inhibition results were compared with control data combined from drug treatments that did not reduce intake, including NMDAR or PKC inhibition in Shell, Ox1R inhibition in accumbens core, and systemic inhibition of dopamine-1 receptors; these were used to understand whether more specific Shell Ox1R contributions in higher drinkers might simply result from intrinsic variability in mouse drinking. Ineffectiveness of Shell inhibition in moderate-drinkers was not due to a floor effect, since systemic baclofen reduced alcohol drinking regardless of basal intake levels, without altering concurrent water intake or saccharin consumption. Finally, alcohol intake in the first exposure predicted consumption levels weeks later, suggesting that intake level may be a stable trait in each individual. Together, our studies indicate that Shell Ox1Rs are critical mediators of binge alcohol intake in higher-drinking individuals, with little net contribution to alcohol drinking in more moderate bingers, and that targeting Ox1Rs may substantially reduce AUD-related harms.
Collapse
Affiliation(s)
- Kelly Lei
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Claudina Kwok
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - David Darevsky
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Scott A Wegner
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - JiHwan Yu
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Lisa Nakayama
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Vincent Pedrozo
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Lexy Anderson
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Shahbaj Ghotra
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Mary Fouad
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Frederic W Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
24
|
Role of glutamatergic system and mesocorticolimbic circuits in alcohol dependence. Prog Neurobiol 2018; 171:32-49. [PMID: 30316901 DOI: 10.1016/j.pneurobio.2018.10.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/08/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
Emerging evidence demonstrates that alcohol dependence is associated with dysregulation of several neurotransmitters. Alterations in dopamine, glutamate and gamma-aminobutyric acid release are linked to chronic alcohol exposure. The effects of alcohol on the glutamatergic system in the mesocorticolimbic areas have been investigated extensively. Several studies have demonstrated dysregulation in the glutamatergic systems in animal models exposed to alcohol. Alcohol exposure can lead to an increase in extracellular glutamate concentrations in mesocorticolimbic brain regions. In addition, alcohol exposure affects the expression and functions of several glutamate receptors and glutamate transporters in these brain regions. In this review, we discussed the effects of alcohol exposure on glutamate receptors, glutamate transporters and glutamate homeostasis in each area of the mesocorticolimbic system. In addition, we discussed the genetic aspect of alcohol associated with glutamate and reward circuitry. We also discussed the potential therapeutic role of glutamate receptors and glutamate transporters in each brain region for the treatment of alcohol dependence. Finally, we provided some limitations on targeting the glutamatergic system for potential therapeutic options for the treatment alcohol use disorders.
Collapse
|
25
|
Newman EL, Albrechet-Souza L, Andrew PM, Auld JG, Burk KC, Hwa LS, Zhang EY, DeBold JF, Miczek KA. Persistent escalation of alcohol consumption by mice exposed to brief episodes of social defeat stress: suppression by CRF-R1 antagonism. Psychopharmacology (Berl) 2018; 235:1807-1820. [PMID: 29696309 PMCID: PMC6168197 DOI: 10.1007/s00213-018-4905-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
RATIONALE Episodic bouts of social stress can precede the initiation, escalation, or relapse to disordered alcohol intake. Social stress may engender neuroadaptations in the hypothalamic-pituitary-adrenal (HPA) axis and in extrahypothalamic stress circuitry to promote the escalation of alcohol intake. OBJECTIVES We aimed to (1) confirm a pattern of escalated drinking in socially defeated mice and to (2) test drugs that target distinct aspects of the HPA axis and extrahypothalamic neural substrates for their effectiveness in reducing murine, stress-escalated drinking. METHODS Male C57BL/6J (B6) mice were socially defeated by resident Swiss-derived males for ten consecutive days receiving 30 bites/day. Ten days after the final defeat, cohorts of B6 mice received continuous or intermittent access to 20% EtOH (w/v) and water. After 4 weeks of drinking, mice were injected with weekly, systemic doses of the CRF-R1 antagonist, CP376395; the glucocorticoid receptor antagonist, mifepristone; the 11-beta-hydroxylase inhibitor, metyrapone; or the 5-alpha-reductase inhibitor, finasteride. RESULTS Prior to drug treatments, defeated mice reliably consumed more EtOH than non-defeated controls, and mice given alcohol intermittently consumed more EtOH than those with continuous access. CP376395 (17-30 mg/kg) reduced continuous, but not intermittent EtOH intake (g/kg) in socially defeated mice. Mifepristone (100 mg/kg), however, increased drinking by defeated mice with intermittent access to alcohol while reducing drinking during continuous access. When administered finasteride (100 mg/kg) or metyrapone (50 mg/kg), all mice reduced their EtOH intake while increasing their water consumption. CONCLUSIONS Mice with a history of episodic social defeat stress were selectively sensitive to the effects of CRF-R1 antagonism, suggesting that CRF-R1 may be a potential target for treating alcohol use disorders in individuals who escalate their drinking after exposure to repeated bouts of psychosocial stress. Future studies will clarify how social defeat stress may alter the expression of extrahypothalamic CRF-R1 and glucocorticoid receptors.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Lucas Albrechet-Souza
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Peter M Andrew
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - John G Auld
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Kelly C Burk
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Lara S Hwa
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Eric Y Zhang
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Joseph F DeBold
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Klaus A Miczek
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA, 02155, USA.
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, 02111, USA.
| |
Collapse
|
26
|
Newman EL, Terunuma M, Wang TL, Hewage N, Bicakci MB, Moss SJ, DeBold JF, Miczek KA. A Role for Prefrontal Cortical NMDA Receptors in Murine Alcohol-Heightened Aggression. Neuropsychopharmacology 2018; 43:1224-1234. [PMID: 29052618 PMCID: PMC5916347 DOI: 10.1038/npp.2017.253] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/03/2017] [Accepted: 10/15/2017] [Indexed: 11/09/2022]
Abstract
Alcohol is associated with nearly half of all violent crimes committed in the United States; yet, a potential neural basis for this type of pathological aggression remains elusive. Alcohol may act on N-methyl-d-aspartate receptors (NMDARs) within cortical circuits to impede processing and to promote aggression. Here, male mice were characterized as alcohol-heightened (AHAs) or alcohol non-heightened aggressors (ANAs) during resident-intruder confrontations after self-administering 1.0 g/kg alcohol (6% w/v) or water. Alcohol produced a pathological-like pattern of aggression in AHAs; these mice shifted their bites to more vulnerable locations on the body of a submissive animal, including the anterior back and ventrum after consuming alcohol. In addition, through immunoblotting, we found that AHAs overexpressed the NMDAR GluN2D subunit in the prefrontal cortex (PFC) as compared to ANAs while the two phenotypes expressed similar levels of GluN1, GluN2A and GluN2B. After identifying several behavioral and molecular characteristics that distinguish AHAs from ANAs, we tested additional mice for their aggression following preferential antagonism of GluN2D-containing NMDARs. In these experiments, groups of AHAs and ANAs self-administered 1.0 g/kg alcohol (6% w/v) or water before receiving intraperitoneal (i.p.) doses of ketamine or memantine, or infusions of memantine directly into the prelimbic (PLmPFC) or infralimbic medial PFC (ILmPFC). Moderate doses of IP ketamine, IP memantine, or intra-PLmPFC memantine increased aggression in AHAs, but only in the absence of alcohol. Prior alcohol intake blocked the pro-aggressive effects of ketamine or memantine. In contrast, only memantine, administered systemically or intra-PLmPFC, interacted with prior alcohol intake to escalate aggression in ANAs. Intra-ILmPFC memantine had no effect on aggression in either AHAs or ANAs. In sum, this work illustrates a potential role of GluN2D-containing NMDARs in the PLmPFC in alcohol-heightened aggression. GluN2D-containing NMDARs are highly expressed on cortical parvalbumin-containing interneurons, suggesting that, in a subset of individuals, alcohol may functionally alter signal integration within cortical microcircuits to dysregulate threat reactivity and promote aggression. This work suggests that targeting GluN2D-NMDARs may be of use in reducing the impact of alcohol-related violence in the human population.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Department, Tufts University, Medford, MA, USA
| | - Miho Terunuma
- Division of Oral Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tiffany L Wang
- Psychology Department, Tufts University, Medford, MA, USA
| | - Nishani Hewage
- Psychology Department, Tufts University, Medford, MA, USA
| | | | - Stephen J Moss
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| | | | - Klaus A Miczek
- Psychology Department, Tufts University, Medford, MA, USA
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| |
Collapse
|
27
|
Kitanaka N, Kitanaka J, Hall FS, Kubota Y, Mimura Y, Ogura S, Okada Y, Uhl GR, Takemura M. Psychotomimetic-like behavioral effects of memantine in the mouse. Biomed Pharmacother 2018; 100:116-123. [PMID: 29427922 DOI: 10.1016/j.biopha.2018.01.160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022] Open
Abstract
A single administration of mice with memantine (1-amino-3,5-dimethyladamantane), a glutamatergic N-methyl-d-aspartate (NMDA) receptor antagonist, induced stereotyped behaviors in dose- and time-dependent manners. The predominant behavioral component of the stereotypy was a continuous, exaggerated sniffing which was accompanied by persistent locomotion. In contrast, a psychostimulant methamphetamine (METH) predominantly induced a stereotyped biting and other forms of intense stationary stereotypical behaviors. Memantine-induced stereotyped sniffing was attenuated by pretreatment with haloperidol, a dopamine D2 receptor antagonist, in a dose-dependent manner. The memantine-induced stereotyped sniffing was also attenuated by pretreatment with betahistine (2-[2-(methylamino)ethyl]pyridine), an agent which increases histamine turnover and releases histamine in the brain. These observations suggest that memantine might induce stereotypies through neuronal mechanisms that are somewhat different from those of METH, but still overlap to a certain extent, since memantine-induced stereotypies can be attenuated by the mechanisms that also suppress METH-induced stereotypy. Importantly, these data suggests that the effects of memantine may be more limited to the ventral striatum including nucleus accumbens than those of METH, which is associated with dorsal striatal stimulation at high doses. In this respect memantine may also have pharmacological properties such as compartmentation (i.e. brain distribution) and neuronal mechanisms different from those of other NMDA receptor antagonists, such as ketamine, which may have important implications for therapeutic uses of these drugs.
Collapse
Affiliation(s)
- Nobue Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Junichi Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan.
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Yoshiro Kubota
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Yumi Mimura
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Sayaka Ogura
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Yukiya Okada
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - George R Uhl
- New Mexico VA Healthcare System/BRINM, Albuquerque, NM 87108, USA
| | - Motohiko Takemura
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| |
Collapse
|
28
|
Cuzon Carlson VC. GABA and Glutamate Synaptic Coadaptations to Chronic Ethanol in the Striatum. Handb Exp Pharmacol 2018; 248:79-112. [PMID: 29460153 DOI: 10.1007/164_2018_98] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Alcohol (ethanol) is a widely used and abused drug with approximately 90% of adults over the age of 18 consuming alcohol at some point in their lifetime. Alcohol exerts its actions through multiple neurotransmitter systems within the brain, most notably the GABAergic and glutamatergic systems. Alcohol's actions on GABAergic and glutamatergic neurotransmission have been suggested to underlie the acute behavioral effects of ethanol. The striatum is the primary input nucleus of the basal ganglia that plays a role in motor and reward systems. The effect of ethanol on GABAergic and glutamatergic neurotransmission within striatal circuitry has been thought to underlie ethanol taking, seeking, withdrawal and relapse. This chapter reviews the effects of ethanol on GABAergic and glutamatergic transmission, highlighting the dynamic changes in striatal circuitry from acute to chronic exposure and withdrawal.
Collapse
|
29
|
Bernardi RE, Broccoli L, Hirth N, Justice NJ, Deussing JM, Hansson AC, Spanagel R. Dissociable Role of Corticotropin Releasing Hormone Receptor Subtype 1 on Dopaminergic and D1 Dopaminoceptive Neurons in Cocaine Seeking Behavior. Front Behav Neurosci 2017; 11:221. [PMID: 29180955 PMCID: PMC5693884 DOI: 10.3389/fnbeh.2017.00221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/24/2017] [Indexed: 11/18/2022] Open
Abstract
The ability of many drugs of abuse, including cocaine, to mediate reinforcement and drug-seeking behaviors is in part mediated by the corticotropin-releasing hormone (CRH) system, in which CRH exerts its effects partly via the CRH receptor subtype 1 (CRHR1) in extra-hypothalamic areas. In fact, CRHR1 expressed in regions of the mesolimbic dopamine (DA) system have been demonstrated to modify cocaine-induced DA release and alter cocaine-mediated behaviors. Here we examined the role of neuronal selectivity of CRHR1 within the mesolimbic system on cocaine-induced behaviors. First we used a transgenic mouse line expressing GFP under the control of the Crhr1 promoter for double fluorescence immunohistochemistry to demonstrate the cellular location of CRHR1 in both dopaminergic and D1 dopaminoceptive neurons. We then studied cocaine sensitization, self-administration, and reinstatement in inducible CRHR1 knockouts using the CreERT2/loxP in either dopamine transporter (DAT)-containing neurons (DAT-Crhr1) or dopamine receptor 1 (D1)-containing neurons (D1-Crhr1). For sensitization testing, mice received five daily injections of cocaine (15 mg/kg IP). For self-administration, mice received eight daily 2 h cocaine (0.5 mg/kg per infusion) self-administration sessions followed by extinction and reinstatement testing. There were no differences in the acute or sensitized locomotor response to cocaine in DAT-Crhr1 or D1-Crhr1 mice and their respective controls. Furthermore, both DAT-Crhr1 and D1-Crhr1 mice reliably self-administered cocaine at the level of controls. However, DAT-Crhr1 mice demonstrated a significant increase in cue-induced reinstatement relative to controls, whereas D1-Crhr1 mice demonstrated a significant decrease in cue-induced reinstatement relative to controls. These data demonstrate the involvement of CRHR1 in cue-induced reinstatement following cocaine self-administration, and implicate a bi-directional role of CRHR1 for cocaine craving.
Collapse
Affiliation(s)
- Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Laura Broccoli
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Natalie Hirth
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas, Houston, TX, United States
| | - Jan M Deussing
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
30
|
Rotermund C, Reolon GK, Leixner S, Boden C, Bilbao A, Kahle PJ. Enhanced motivation to alcohol in transgenic mice expressing human α-synuclein. J Neurochem 2017; 143:294-305. [PMID: 28833174 DOI: 10.1111/jnc.14151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/20/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022]
Abstract
α-Synuclein (αSYN) is the neuropathological hallmark protein of Parkinson's disease (PD) and related neurodegenerative disorders. Moreover, the gene encoding αSYN (SNCA) is a major genetic contributor to PD. Interestingly, independent genome-wide association studies also identified SNCA as the most important candidate gene for alcoholism. Furthermore, single-nucleotide-polymorphisms have been associated with alcohol-craving behavior and alcohol-craving patients showed augmented αSYN expression in blood. To investigate the effect of αSYN on the addictive properties of chronic alcohol use, we examined consumption, motivation, and seeking responses induced by environmental stimuli and relapse behavior in transgenic mice expressing the human mutant [A30P]αSYN throughout the brain. The primary reinforcing effects of alcohol under operant self-administration conditions were increased, while consumption and the alcohol deprivation effect were not altered in the transgenic mice. The same mice were subjected to immunohistochemical measurements of immediate-early gene inductions in brain regions involved in addiction-related behaviors. Acute ethanol injection enhanced immunostaining for the phosphorylated form of cAMP response element binding protein in both amygdala and nucleus accumbens of αSYN transgenic mice, while in wild-type mice no effect was visible. However, at the same time, levels of cFos remain unchanged in both genotypes. These results provide experimental confirmation of SNCA as a candidate gene for alcoholism in addition to its known link to PD.
Collapse
Affiliation(s)
- Carola Rotermund
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany
| | - Gustavo K Reolon
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute of Clinical Brain Research, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Sarah Leixner
- Behavioral Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Cindy Boden
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany
| | - Ainhoa Bilbao
- Behavioral Genetics Research Group, Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Philipp J Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, German Center of Neurodegenerative Diseases, Tübingen, Germany.,Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute of Clinical Brain Research, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| |
Collapse
|
31
|
Fudalej S, Klimkiewicz A, Mach A, Jakubczyk A, Fudalej M, Wasilewska K, Podgórska A, Krajewski P, Płoski R, Wojnar M. An association between genetic variation in the glutamatergic system and suicide attempts in alcohol-dependent individuals. Am J Addict 2017; 26:595-601. [DOI: 10.1111/ajad.12571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/19/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Affiliation(s)
- Sylwia Fudalej
- Department of Psychiatry; Medical University of Warsaw; Warsaw Poland
| | - Anna Klimkiewicz
- Department of Psychiatry; Medical University of Warsaw; Warsaw Poland
| | - Anna Mach
- Department of Psychiatry; Medical University of Warsaw; Warsaw Poland
| | - Andrzej Jakubczyk
- Department of Psychiatry; Medical University of Warsaw; Warsaw Poland
| | - Marcin Fudalej
- Department of Forensic Medicine; Medical University of Warsaw; Warsaw Poland
| | | | - Anna Podgórska
- The Institute of Physiology and Pathology of Hearing; Kajetany Poland
| | - Paweł Krajewski
- Department of Forensic Medicine; Medical University of Warsaw; Warsaw Poland
| | - Rafał Płoski
- Department of Medical Genetics; Medical University of Warsaw; Warsaw Poland
| | - Marcin Wojnar
- Department of Psychiatry; Medical University of Warsaw; Warsaw Poland
- Department of Psychiatry; University of Michigan; Ann Arbor Michigan
| |
Collapse
|
32
|
Egervari G, Landry J, Callens J, Fullard JF, Roussos P, Keller E, Hurd YL. Striatal H3K27 Acetylation Linked to Glutamatergic Gene Dysregulation in Human Heroin Abusers Holds Promise as Therapeutic Target. Biol Psychiatry 2017; 81:585-594. [PMID: 27863698 PMCID: PMC5346335 DOI: 10.1016/j.biopsych.2016.09.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Opiate abuse and overdose reached epidemic levels in the United States. However, despite significant advances in animal and in vitro models, little knowledge has been directly accrued regarding the neurobiology of the opiate-addicted human brain. METHODS We used postmortem human brain specimens from a homogeneous European Caucasian population of heroin users for transcriptional and epigenetic profiling, as well as direct assessment of chromatin accessibility in the striatum, a brain region central to reward and emotion. A rat heroin self-administration model was used to obtain translational molecular and behavioral insights. RESULTS Our transcriptome approach revealed marked impairments related to glutamatergic neurotransmission and chromatin remodeling in the human striatum. A series of biochemical experiments tracked the specific location of the epigenetic disturbances to hyperacetylation of lysine 27 of histone H3, showing dynamic correlations with heroin use history and acute opiate toxicology. Targeted investigation of GRIA1, a glutamatergic gene implicated in drug-seeking behavior, verified the increased enrichment of lysine-27 acetylated histone H3 at discrete loci, accompanied by enhanced chromatin accessibility at hyperacetylated regions in the gene body. Analogous epigenetic impairments were detected in the striatum of heroin self-administering rats. Using this translational model, we showed that bromodomain inhibitor JQ1, which blocks the functional readout of acetylated lysines, reduced heroin self-administration and cue-induced drug-seeking behavior. CONCLUSIONS Overall, our data suggest that heroin-related histone H3 hyperacetylation contributes to glutamatergic transcriptional changes that underlie addiction behavior and identify JQ1 as a promising candidate for targeted clinical interventions in heroin use disorder.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Psychiatry, Friedman Brain Institute; Fishberg Department of Neuroscience, Friedman Brain Institute
| | - Joseph Landry
- Department of Psychiatry, Friedman Brain Institute; Fishberg Department of Neuroscience, Friedman Brain Institute
| | - James Callens
- Department of Psychiatry, Friedman Brain Institute; Fishberg Department of Neuroscience, Friedman Brain Institute
| | - John F Fullard
- Department of Psychiatry, Friedman Brain Institute; Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York
| | - Panos Roussos
- Department of Psychiatry, Friedman Brain Institute; Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York; Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, New York
| | - Eva Keller
- Department of Forensic Medicine, Semmelweis University, Budapest, Hungary
| | - Yasmin L Hurd
- Department of Psychiatry, Friedman Brain Institute; Fishberg Department of Neuroscience, Friedman Brain Institute.
| |
Collapse
|
33
|
Goodwani S, Saternos H, Alasmari F, Sari Y. Metabotropic and ionotropic glutamate receptors as potential targets for the treatment of alcohol use disorder. Neurosci Biobehav Rev 2017; 77:14-31. [PMID: 28242339 DOI: 10.1016/j.neubiorev.2017.02.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicates that dysfunctional glutamate neurotransmission is critical in the initiation and development of alcohol and drug dependence. Alcohol consumption induced downregulation of glutamate transporter 1 (GLT-1) as reported in previous studies from our laboratory. Glutamate is the major excitatory neurotransmitter in the brain, which acts via interactions with several glutamate receptors. Alcohol consumption interferes with the glutamatergic signal transmission by altering the functions of these receptors. Among the glutamate receptors involved in alcohol-drinking behavior are the metabotropic receptors such as mGluR1/5, mGluR2/3, and mGluR7, as well as the ionotropic receptors, NMDA and AMPA. Preclinical studies using agonists and antagonists implicate these glutamatergic receptors in the development of alcohol use disorder (AUD). Therefore, the purpose of this review is to discuss the neurocircuitry involving glutamate transmission in animals exposed to alcohol and further outline the role of metabotropic and ionotropic receptors in the regulation of alcohol-drinking behavior. This review provides ample information about the potential therapeutic role of glutamatergic receptors for the treatment of AUD.
Collapse
Affiliation(s)
- Sunil Goodwani
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA; The Neurodegeneration Consortium, Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Hannah Saternos
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA
| | - Fawaz Alasmari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA
| | - Youssef Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA.
| |
Collapse
|
34
|
Hopf FW. Do specific NMDA receptor subunits act as gateways for addictive behaviors? GENES BRAIN AND BEHAVIOR 2016; 16:118-138. [PMID: 27706932 DOI: 10.1111/gbb.12348] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 12/19/2022]
Abstract
Addiction to alcohol and drugs is a major social and economic problem, and there is considerable interest in understanding the molecular mechanisms that promote addictive drives. A number of proteins have been identified that contribute to expression of addictive behaviors. NMDA receptors (NMDARs), a subclass of ionotropic glutamate receptors, have been of particular interest because their physiological properties make them an attractive candidate for gating induction of synaptic plasticity, a molecular change thought to mediate learning and memory. NMDARs are generally inactive at the hyperpolarized resting potentials of many neurons. However, given sufficient depolarization, NMDARs are activated and exhibit long-lasting currents with significant calcium permeability. Also, in addition to stimulating neurons by direct depolarization, NMDARs and their calcium signaling can allow strong and/or synchronized inputs to produce long-term changes in other molecules (such as AMPA-type glutamate receptors) which can last from days to years, binding internal and external stimuli in a long-term memory trace. Such memories could allow salient drug-related stimuli to exert strong control over future behaviors and thus promote addictive drives. Finally, NMDARs may themselves undergo plasticity, which can alter subsequent neuronal stimulation and/or the ability to induce plasticity. This review will address recent and past findings suggesting that NMDAR activity promotes drug- and alcohol-related behaviors, with a particular focus on GluN2B subunits as possible central regulators of many addictive behaviors, as well as newer studies examining the importance of non-canonical NMDAR subunits and endogenous NMDAR cofactors.
Collapse
Affiliation(s)
- F W Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
35
|
Metabolic shift of the kynurenine pathway impairs alcohol and cocaine seeking and relapse. Psychopharmacology (Berl) 2016; 233:3449-59. [PMID: 27475106 DOI: 10.1007/s00213-016-4384-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/16/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE The glutamatergic system plays a key role in the maintenance of drug use and development of drug-related conditioned behaviours. In particular, hyper-glutamatergic activity and N-methyl-D-aspartate receptor (NMDAR) activation may drive drug craving and relapse. Inhibition of kynurenine-3-monooxygenase (KMO) shifts the metabolic kynurenine pathway towards production of kynurenic acid, which leads to a reduction of glutamatergic/NMDAR activity via different mechanisms. OBJECTIVES In this study, we investigated whether drug-seeking and relapse behaviour could be modified by the metabolic shift of endogenous kynurenine pathway. METHODS An inhibitor of kynurenine-3-monooxygenase (KMO) Ro61-8048 (4 and 40 mg/kg) and its prodrug JM6 (100 and 200 mg/kg) were tested in two behavioural rat models for drug seeking and relapse-the alcohol deprivation effect (ADE) model in long-term alcohol-drinking rats and the model of cue-induced reinstatement of alcohol- and cocaine-seeking behaviour. RESULTS Our results show that relapse-like alcohol drinking during the ADE was abolished by repeated intraperitoneal administration of Ro61-8048 and significantly reduced by its oral prodrug JM6. Cue-induced reinstatement of both alcohol- and cocaine-seeking behaviour was also abolished by administration of Ro61-8048. CONCLUSIONS Pharmacological enhancement of endogenous kynurenic acid levels provides a novel treatment strategy to interfere with glutamatergic/NMDAR activity as well as with craving and relapse in alcohol-dependent patients and drug addicts.
Collapse
|
36
|
Pina MM, Cunningham CL. Involvement of ventral tegmental area ionotropic glutamate receptors in the expression of ethanol-induced conditioned place preference. Behav Brain Res 2016; 313:23-29. [PMID: 27378337 DOI: 10.1016/j.bbr.2016.06.063] [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: 04/08/2016] [Revised: 06/16/2016] [Accepted: 06/30/2016] [Indexed: 01/13/2023]
Abstract
The ventral tegmental area (VTA) is a well-established neural substrate of reward-related processes. Activity within this structure is increased by the primary and conditioned rewarding effects of abused drugs and its engagement is heavily reliant on excitatory input from structures upstream. In the case of drug seeking, it is thought that exposure to drug-associated cues engages glutamatergic VTA afferents that signal directly to dopamine cells, thereby triggering this behavior. It is unclear, however, whether glutamate input to VTA is directly involved in ethanol-associated cue seeking. Here, the role of intra-VTA ionotropic glutamate receptor (iGluR) signaling in ethanol-cue seeking was evaluated in DBA/2J mice using an ethanol conditioned place preference (CPP) procedure. Intra-VTA iGluRs α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPAR)/kainate and N-methyl-d-aspartate (NMDAR) were blocked during ethanol CPP expression by co-infusion of antagonist drugs 6,7-dinitroquinoxaline-2,3-dione (DNQX; AMPA/kainate) and d-(-)-2-Amino-5-phosphonopentanoic acid (AP5; NMDA). Compared to aCSF, bilateral infusion of low (1 DNQX+100 AP5ng/side) and high (5 DNQX+500 AP5ng/side) doses of the AMPAR and NMDAR antagonist cocktail into VTA blocked ethanol CPP expression. This effect was site specific, as DNQX/AP5 infusion proximal to VTA did not significantly impact CPP expression. An increase in activity was found at the high but not low dose of DNQX/AP5. These findings demonstrate that activation of iGluRs within the VTA is necessary for ethanol-associated cue seeking, as measured by CPP.
Collapse
Affiliation(s)
- Melanie M Pina
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239-3098, USA.
| | - Christopher L Cunningham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239-3098, USA
| |
Collapse
|
37
|
Sathler MF, Stutz B, Martins RS, Dos Santos Pereira M, Pecinalli NR, Santos LE, Taveira-da-Silva R, Lowe J, de Freitas IG, de Melo Reis RA, Manhães AC, Kubrusly RCC. Single exposure to cocaine impairs aspartate uptake in the pre-frontal cortex via dopamine D1-receptor dependent mechanisms. Neuroscience 2016; 329:326-36. [PMID: 27208619 DOI: 10.1016/j.neuroscience.2016.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 11/16/2022]
Abstract
Dopamine and glutamate play critical roles in the reinforcing effects of cocaine. We demonstrated that a single intraperitoneal administration of cocaine induces a significant decrease in [(3)H]-d-aspartate uptake in the pre-frontal cortex (PFC). This decrease is associated with elevated dopamine levels, and requires dopamine D1-receptor signaling (D1R) and adenylyl cyclase activation. The effect was observed within 10min of cocaine administration and lasted for up to 30min. This rapid response is related to D1R-mediated cAMP-mediated activation of PKA and phosphorylation of the excitatory amino acid transporters EAAT1, EAAT2 and EAAT3. We also demonstrated that cocaine exposure increases extracellular d-aspartate, l-glutamate and d-serine in the PFC. Our data suggest that cocaine activates dopamine D1 receptor signaling and PKA pathway to regulate EAATs function and extracellular EAA level in the PFC.
Collapse
Affiliation(s)
- Matheus Figueiredo Sathler
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Bernardo Stutz
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Robertta Silva Martins
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Maurício Dos Santos Pereira
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil; Laboratório de Neurofisiologia Molecular, Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| | - Ney Roner Pecinalli
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Luis E Santos
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Rosilane Taveira-da-Silva
- Laboratório de Fisico-Química Biológica Aída Hassón-Voloch Instituto de Biofísica Carlos Chagas Filho Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Jennifer Lowe
- Laboratório de Fisico-Química Biológica Aída Hassón-Voloch Instituto de Biofísica Carlos Chagas Filho Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Isis Grigorio de Freitas
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Regina C C Kubrusly
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|