1
|
Peyton L, Haroon H, Umpierre A, Essa H, Bruce R, Wu LJ, Choi DS. In vivo calcium extrusion from accumbal astrocytes reduces anxiety-like behaviors but increases compulsive-like responses and compulsive ethanol drinking in mice. Neuropharmacology 2025; 268:110320. [PMID: 39842625 PMCID: PMC11830519 DOI: 10.1016/j.neuropharm.2025.110320] [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: 12/22/2023] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
The ventral striatum is crucially involved in reward processing. The present study investigates the behavioral effects of astrocyte-specific calcium extrusion virus "CalEx" on perseverative responses in the operant five-choice serial reaction time task and ethanol-conditioned place preference. Mice were injected with CalEx via the GfaABC1D promoter to extrude cytosolic calcium from astrocytes within the ventral striatum. We found that CalEx transfection in the ventral striatum reduced evoked response duration, the maximum amplitude, and the response frequency to 500 μM ATP as measured by ΔF/F fluorescence intensity of the genetically encoded calcium indicator targeting astrocytes GCaMP6f. During the five-choice serial reaction time task, CalEx mice persisted in perseverative responses compared to their counterparts. Additionally, during ethanol-conditioned place preference, CalEx mice showed increased place preference for a low ethanol concentration compared to control group. Furthermore, we found that accumbal astrocytic calcium extrusion increased quinine adulterated ethanol drinking. Our findings suggest that diminishing ventral striatum astrocyte calcium activity contributes to compulsive behaviors, ethanol drinking, and enhanced ethanol drug reward.
Collapse
Affiliation(s)
- Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Humza Haroon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | | | - Hesham Essa
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Robert Bruce
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 59905, USA.
| |
Collapse
|
2
|
Goyal A, Cabrera JR, Blaha CD, Lee KH, Shin H, Oh Y. Ventral tegmental area deep brain stimulation reverses ethanol-induced dopamine increase in the rat nucleus accumbens. Biomed Eng Lett 2024; 14:1347-1354. [PMID: 39465114 PMCID: PMC11502691 DOI: 10.1007/s13534-024-00408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/09/2024] [Accepted: 06/30/2024] [Indexed: 10/29/2024] Open
Abstract
The neurophysiology of alcohol use disorder (AUD) is complex, but a major contributor to addictive phenotypes is the tendency for drugs of abuse to increase tonic extracellular dopamine (DA) levels in the nucleus accumbens (NAc). Repeated exposure to substances of abuse such as ethanol results in the overstimulation of the mesolimbic pathway, causing an excessive release of DA from the ventral tegmental area (VTA) to target regions such as the NAc. This heightened DA signaling is associated with the reinforcing effects of substances, leading to a strong desire for continued use. Recent work has postulated that high frequency deep brain stimulation (DBS) of the ventral tegmental area may reduce dopamine transmission to the nucleus accumbens following acute drug of abuse exposure, thereby mitigating the drug's addictive potential. We first demonstrate ethanol's ability to decrease phasic DA release over time and to increase tonic extracellular DA concentrations in the nucleus accumbens. Next, we demonstrate the capability for high frequency VTA DBS to reverse this ethanol-associated surge in tonic DA concentrations in the nucleus accumbens to levels not significantly different from baseline. This study suggests a promising new avenue for investigating the mechanisms of alcohol use disorder.
Collapse
Affiliation(s)
- Abhinav Goyal
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905 USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Juan Rojas Cabrera
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905 USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Charles D. Blaha
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Kendall H. Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| | - Hojin Shin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| | - Yoonbae Oh
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
3
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in dopamine neurons promotes EtOH seeking, behavioral reward, and susceptibility to relapse. Psychopharmacology (Berl) 2024; 241:2255-2269. [PMID: 38856764 PMCID: PMC11518640 DOI: 10.1007/s00213-024-06631-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). The neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6fl/fl; DAT-iCreER mice to determine the role of RGS6 in DA neurons on EtOH consumption, reward, and relapse behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse ventral tegmental area (VTA), and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6-/- mice. Strikingly, RGS6 loss from DA neurons before or after EtOH behavioral reward is established significantly reduced (~ 50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies identify DA neurons as the locus of RGS6 action in promoting EtOH consumption, preference, reward, and relapse. RGS6 is unique among R7 RGS proteins in promoting rather than suppressing behavioral responses to drugs of abuse and to modulate EtOH behavioral reward. This is a result of RGS6's pre-synaptic actions that we hypothesize promote VTA DA transmission by suppressing GPCR-Gαi/o-DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
Affiliation(s)
- Mackenzie M Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Matthew A Weber
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
| | - Nandakumar S Narayanan
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Rory A Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA.
| |
Collapse
|
4
|
Bassareo V, Maccioni R, Talani G, Zuffa S, El Abiead Y, Lorrai I, Kawamura T, Pantis S, Puliga R, Vargiu R, Lecca D, Enrico P, Peana A, Dazzi L, Dorrestein PC, Sanna PP, Sanna E, Acquas E. Receptor and metabolic insights on the ability of caffeine to prevent alcohol-induced stimulation of mesolimbic dopamine transmission. Transl Psychiatry 2024; 14:391. [PMID: 39341817 PMCID: PMC11438888 DOI: 10.1038/s41398-024-03112-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
The consumption of alcohol and caffeine affects the lives of billions of individuals worldwide. Although recent evidence indicates that caffeine impairs the reinforcing properties of alcohol, a characterization of its effects on alcohol-stimulated mesolimbic dopamine (DA) function was lacking. Acting as the pro-drug of salsolinol, alcohol excites DA neurons in the posterior ventral tegmental area (pVTA) and increases DA release in the nucleus accumbens shell (AcbSh). Here we show that caffeine, via antagonistic activity on A2A adenosine receptors (A2AR), prevents alcohol-dependent activation of mesolimbic DA function as assessed, in-vivo, by brain microdialysis of AcbSh DA and, in-vitro, by electrophysiological recordings of pVTA DA neuronal firing. Accordingly, while the A1R antagonist DPCPX fails to prevent the effects of alcohol on DA function, both caffeine and the A2AR antagonist SCH 58261 prevent alcohol-dependent pVTA generation of salsolinol and increase in AcbSh DA in-vivo, as well as alcohol-dependent excitation of pVTA DA neurons in-vitro. However, caffeine also prevents direct salsolinol- and morphine-stimulated DA function, suggesting that it can exert these inhibitory effects also independently from affecting alcohol-induced salsolinol formation or bioavailability. Finally, untargeted metabolomics of the pVTA showcases that caffeine antagonizes alcohol-mediated effects on molecules (e.g. phosphatidylcholines, fatty amides, carnitines) involved in lipid signaling and energy metabolism, which could represent an additional salsolinol-independent mechanism of caffeine in impairing alcohol-mediated stimulation of mesolimbic DA transmission. In conclusion, the outcomes of this study strengthen the potential of caffeine, as well as of A2AR antagonists, for future development of preventive/therapeutic strategies for alcohol use disorder.
Collapse
Affiliation(s)
- Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Riccardo Maccioni
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Giuseppe Talani
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Irene Lorrai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoya Kawamura
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sofia Pantis
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta Puliga
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Romina Vargiu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Daniele Lecca
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Paolo Enrico
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandra Peana
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Laura Dazzi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Enrico Sanna
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| |
Collapse
|
5
|
Song RX, Miao HT, Jia SY, Li WG, Liu JZ, Zhang W, Xing BR, Zhao JY, Zhang LM, Li XM. Hemorrhagic Shock and Resuscitation Causes Excessive Dopaminergic Signaling in the mPFC and Cognitive Dysfunction. Mol Neurobiol 2024; 61:3899-3910. [PMID: 38041715 DOI: 10.1007/s12035-023-03804-y] [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: 08/22/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
Peri-operative hemorrhagic shock and resuscitation (HSR), a severe traumatic stress, is closely associated with post-operative anxiety, depression, and cognitive dysfunction, subsequently causing a serious burden on families and society. Following the co-release of corticotropin-releasing factor and catecholamine, traumatic stress activates dopaminergic neurons, increasing the addictive behavior and neurocognitive impairment risks. This study investigates the association between cognitive dysfunction and dopaminergic neurons in the mPFC under HSR conditions. This study established an HSR model by bleeding and re-transfusion in the mice. After HSR exposure, a dopamine D1 receptor antagonist, SKF-83566, was administered intraperitoneally for three consecutive days. Novel object recognition (NOR), conditioned fearing (FC), and conditioned place preference (CPP) were used to assess cognitive changes 16 days after HSR exposure. Local field potential (LFP) in the mPFC was also investigated during the novel object exploration. Compared with the mice exposed to sham, there was a significant decrease in the object recognition index, a reduction in context- and tone-related freezing time, an increase in CPP values, a downregulation of β-power but upregulation of γ-power in the mPFC in the mice exposed to HSR. Moreover, the mice exposed to HSR showed significantly upregulated TH-positive cell number, cleaved caspase-1- and TH-positive cells, and interleukin (IL)-1β/18 expression in the mPFC compared with sham; SKF-83566 could partially reverse these alternations. The HSR caused excessive dopaminergic signaling and cognitive dysfunction in the mPFC, a condition that might be ameliorated using a dopamine D1 receptor inhibitor.
Collapse
Affiliation(s)
- Rong-Xin Song
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Hui-Tao Miao
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Shi-Yan Jia
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Wen-Guang Li
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Ji-Zhen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao-Rui Xing
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Jian-Yong Zhao
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Li-Min Zhang
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China.
| | - Xiao-Ming Li
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China.
| |
Collapse
|
6
|
Maccioni R, Bassareo V, Talani G, Zuffa S, El Abiead Y, Lorrai I, Kawamura T, Pantis S, Puliga R, Vargiu R, Lecca D, Enrico P, Peana A, Dazzi L, Dorrestein PC, Sanna PP, Sanna E, Acquas E. Receptor and metabolic insights on the ability of caffeine to prevent alcohol-induced stimulation of mesolimbic dopamine transmission. RESEARCH SQUARE 2024:rs.3.rs-4289552. [PMID: 38946995 PMCID: PMC11213171 DOI: 10.21203/rs.3.rs-4289552/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The consumption of alcohol and caffeine affects the lives of billions of individuals worldwide. Although recent evidence indicates that caffeine impairs the reinforcing properties of alcohol, a characterization of its effects on alcohol-stimulated mesolimbic dopamine (DA) function was lacking. Acting as the pro-drug of salsolinol, alcohol excites DA neurons in the posterior ventral tegmental area (pVTA) and increases DA release in the nucleus accumbens shell (AcbSh). Here we show that caffeine, via antagonistic activity on A2A adenosine receptors (A2AR), prevents alcohol-dependent activation of mesolimbic DA function as assessed, in-vivo, by brain microdialysis of AcbSh DA and, in-vitro, by electrophysiological recordings of pVTA DA neuronal firing. Accordingly, while the A1R antagonist DPCPX fails to prevent the effects of alcohol on DA function, both caffeine and the A2AR antagonist SCH 58261 prevent alcohol-dependent pVTA generation of salsolinol and increase in AcbSh DA in-vivo, as well as alcohol-dependent excitation of pVTA DA neurons in-vitro. However, caffeine also prevents direct salsolinol- and morphine-stimulated DA function, suggesting that it can exert these inhibitory effects also independently from affecting alcohol-induced salsolinol formation or bioavailability. Finally, untargeted metabolomics of the pVTA showcases that caffeine antagonizes alcohol-mediated effects on molecules (e.g. phosphatidylcholines, fatty amides, carnitines) involved in lipid signaling and energy metabolism, which could represent an additional salsolinol-independent mechanism of caffeine in impairing alcohol-mediated stimulation of mesolimbic DA transmission. In conclusion, the outcomes of this study strengthen the potential of caffeine, as well as of A2AR antagonists, for future development of preventive/therapeutic strategies for alcohol use disorder.
Collapse
|
7
|
Söderpalm B, Ericson M. Alcohol and the dopamine system. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:21-73. [PMID: 38555117 DOI: 10.1016/bs.irn.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The mesolimbic dopamine pathway plays a major role in drug reinforcement and is likely involved also in the development of drug addiction. Ethanol, like most addictive drugs, acutely activates the mesolimbic dopamine system and releases dopamine, and ethanol-associated stimuli also appear to trigger dopamine release. In addition, chronic exposure to ethanol reduces the baseline function of the mesolimbic dopamine system. The molecular mechanisms underlying ethanol´s interaction with this system remain, however, to be unveiled. Here research on the actions of ethanol in the mesolimbic dopamine system, focusing on the involvement of cystein-loop ligand-gated ion channels, opiate receptors, gastric peptides and acetaldehyde is briefly reviewed. In summary, a great complexity as regards ethanol´s mechanism(s) of action along the mesolimbic dopamine system has been revealed. Consequently, several new targets and possibilities for pharmacotherapies for alcohol use disorder have emerged.
Collapse
Affiliation(s)
- Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Dazzi L, Sanna F, Talani G, Bassareo V, Biggio F, Follesa P, Pisu MG, Porcu P, Puliga R, Quartu M, Serra M, Serra MP, Sanna E, Acquas E. Binge-like administration of alcohol mixed to energy drinks to male adolescent rats severely impacts on mesocortical dopaminergic function in adulthood: A behavioral, neurochemical and electrophysiological study. Neuropharmacology 2024; 243:109786. [PMID: 37952712 DOI: 10.1016/j.neuropharm.2023.109786] [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: 06/28/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023]
Abstract
A growing body of evidence indicates that the practice of consuming alcohol mixed with energy drinks (ED) (AMED) in a binge drinking pattern is significantly diffusing among the adolescent population. This behavior, aimed at increasing the intake of alcohol, raises serious concerns about its long-term effects. Epidemiological studies suggest that AMED consumption might increase vulnerability to alcohol abuse and have a gating effect on the use of illicit drugs. The medial prefrontal cortex (mPFC) is involved in the modulation of the reinforcing effects of alcohol and of impulsive behavior and plays a key role in the development of addiction. In our study, we used a binge-like protocol of administration of alcohol, ED, or AMED in male adolescent rats, to mimic the binge-like intake behavior observed in humans, in order to evaluate whether these treatments could differentially affect the function of mesocortical dopaminergic neurons in adulthood. We did so by measuring: i) physiological sensorimotor gating; ii) voluntary alcohol consumption and dopamine transmission before, during, and after presentation of alcohol; iii) electrophysiological activity of VTA dopaminergic neurons and their sensitivity to a challenge with alcohol. Our results indicate that exposure to alcohol, ED, or AMED during adolescence induces differential adaptive changes in the function of mesocortical dopaminergic neurons and, in particular, that AMED exposure decreases their sensitivity to external stimuli, possibly laying the foundation for the altered behaviors observed in adulthood.
Collapse
Affiliation(s)
- Laura Dazzi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Giuseppe Talani
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Francesca Biggio
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Paolo Follesa
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Maria Giuseppina Pisu
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Patrizia Porcu
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Roberta Puliga
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Marina Quartu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Mariangela Serra
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Maria Pina Serra
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| | - Enrico Sanna
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy; Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy.
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, SS 554 - bivio per Sestu, 09042, Monserrato, Cagliari, Italy
| |
Collapse
|
9
|
Chapp AD, Shan Z, Chen QH. Acetic Acid: An Underestimated Metabolite in Ethanol-Induced Changes in Regulating Cardiovascular Function. Antioxidants (Basel) 2024; 13:139. [PMID: 38397737 PMCID: PMC10886048 DOI: 10.3390/antiox13020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Acetic acid is a bioactive short-chain fatty acid produced in large quantities from ethanol metabolism. In this review, we describe how acetic acid/acetate generates oxidative stress, alters the function of pre-sympathetic neurons, and can potentially influence cardiovascular function in both humans and rodents after ethanol consumption. Our recent findings from in vivo and in vitro studies support the notion that administration of acetic acid/acetate generates oxidative stress and increases sympathetic outflow, leading to alterations in arterial blood pressure. Real-time investigation of how ethanol and acetic acid/acetate modulate neural control of cardiovascular function can be conducted by microinjecting compounds into autonomic control centers of the brain and measuring changes in peripheral sympathetic nerve activity and blood pressure in response to these compounds.
Collapse
Affiliation(s)
- Andrew D. Chapp
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhiying Shan
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| | - Qing-Hui Chen
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| |
Collapse
|
10
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in ventral tegmental area (VTA) dopamine neurons promotes EtOH seeking, behavioral reward and susceptibility to relapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563844. [PMID: 37961154 PMCID: PMC10634791 DOI: 10.1101/2023.10.24.563844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). EtOH is the most abused substance worldwide with chronic consumption often leading to the development of dependence and abuse. Unfortunately, the neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6 fl/fl ; DAT-iCreER mice to determine the role of RGS6 in VTA DA neurons on EtOH consumption and reward behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse VTA, and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in VTA DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6 -/- mice. Strikingly, RGS6 loss from VTA DA neurons before or after EtOH behavioral reward is established significantly reduced (∼50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies illuminate a critical role of RGS6 in the mesolimbic circuit in promoting EtOH seeking, reward, and reinstatement. We propose that RGS6 functions to promote DA transmission through its function as a negative modulator of GPCR-Gα i/o -DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
|
11
|
Chapp AD, Collins AR, Driscoll KM, Behnke JE, Shan Z, Zhang L, Chen QH. Ethanol Metabolite, Acetate, Increases Excitability of the Central Nucleus of Amygdala Neurons through Activation of NMDA Receptors. ACS Chem Neurosci 2023; 14:1278-1290. [PMID: 36957993 PMCID: PMC11163875 DOI: 10.1021/acschemneuro.2c00784] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
Abstract
The central nucleus of the amygdala (CeA) is a key brain region involved in emotional and stressor responses due to its many projections to autonomic regulatory centers. It is also a primary site of action from ethanol consumption. However, the influence of active metabolites of ethanol such as acetate on the CeA neural circuitry has yet to be elucidated. Here, we investigated the effect of acetate on CeA neurons with the axon projecting to the rostral ventrolateral medulla (CeA-RVLM), as well as quantified cytosolic calcium responses in primary neuronal cultures. Whole-cell patch-clamp recordings in brain slices containing autonomic CeA-RVLM neurons revealed a dose-dependent increase in neuronal excitability in response to acetate. N-Methyl-d-aspartate receptor (NMDAR) antagonists suppressed the acetate-induced increase in CeA-RVLM neuronal excitability and memantine suppressed the direct activation of NMDAR-dependent inward currents by acetate in brain slices. We observed that acetate increased cytosolic Ca2+ in a time-dependent manner in primary neuronal cell cultures. The acetate enhancement of calcium signaling was abolished by memantine. Computational modeling of acetic acid at NMDAR/NR1 glutamatergic and glycinergic sites suggests potential active site interactions. These findings suggest that within the CeA, acetate is excitatory at least partially through activation of NMDAR, which may underlie the impact of ethanol consumption on autonomic circuitry.
Collapse
Affiliation(s)
- Andrew D Chapp
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
- Department of Neuroscience, University of Minnesota, Twin Cities, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Andréa R Collins
- Department of Psychiatry, University of California, San Francisco, Fresno, California 93701, United States
| | - Kyle M Driscoll
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Jessica E Behnke
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC, 1400 Townsend Drive, Houghton, Michigan 49931, United States
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
12
|
Age-dependent alterations in key components of the nigrostriatal dopaminergic system and distinct motor phenotypes. Acta Pharmacol Sin 2022; 43:862-875. [PMID: 34244603 PMCID: PMC8975991 DOI: 10.1038/s41401-021-00713-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023]
Abstract
The nigrostriatal dopaminergic (DA) system, which includes DA neurons in the ventral and dorsal tiers of the substantia nigra pars compacta (vSNc, dSNc) and DA terminals in the dorsal striatum, is critically implicated in motor control. Accumulating studies demonstrate that both the nigrostriatal DA system and motor function are impaired in aged subjects. However, it is unknown whether dSNc and vSNc DA neurons and striatal DA terminals age in similar patterns, and whether these changes parallel motor deficits. To address this, we performed ex vivo patch-clamp recordings in dSNc and vSNc DA neurons, measured striatal dopamine release, and analyzed motor behaviors in rodents. Spontaneous firing in dSNc and vSNc DA neurons and depolarization-evoked firing in dSNc DA neurons showed inverse V-shaped changes with age. But depolarization-evoked firing in vSNc DA neurons increased with age. In the dorsal striatum, dopamine release declined with age. In locomotor tests, 12-month-old rodents showed hyperactive exploration, relative to 6- and 24-month-old rodents. Additionally, aged rodents showed significant deficits in coordination. Elevating dopamine levels with a dopamine transporter inhibitor improved both locomotion and coordination. Therefore, key components in the nigrostriatal DA system exhibit distinct aging patterns and may contribute to age-related alterations in locomotion and coordination.
Collapse
|
13
|
Robinson JD, Cui Y, Karam-Hage M, Kypriotakis G, Versace F, Ait-Daoud Tiouririne N, Anthenelli RM, Cinciripini PM. Topiramate decreases the salience of motivationally relevant visual cues among smokers with alcohol use disorder. Alcohol Clin Exp Res 2022; 46:384-395. [PMID: 35037278 PMCID: PMC8920769 DOI: 10.1111/acer.14771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND There is preliminary evidence that the anticonvulsant topiramate increases the likelihood of both smoking and alcohol abstinence among smokers with alcohol use disorder (AUD), but its therapeutic mechanism has not been determined. We used event-related potentials (ERPs) to evaluate topiramate's effect on the salience of drug-related, emotional, and neutral pictorial cues to identify whether one of its potential therapeutic mechanisms involves reduction of the salience of motivationally relevant cues. METHODS Participants enrolled in a multisite clinical trial treating smokers with AUD were randomly assigned to receive placebo, low-dose topiramate (up to 125 mg/day), or high-dose topiramate (up to 250 mg/day), along with brief behavioral compliance enhancement treatment. A subsample (n = 101) completed ERP assessments at baseline (1 week pre-medication) and week 5 (5 weeks on medication; 1 week pre-quit). We assessed the salience of pleasant, unpleasant, cigarette-related, alcohol-related, and neutral pictorial cues using the late positive potential (LPP) ERP component and measured self-reported substance use, reinforcement, craving, and withdrawal. RESULTS Five weeks of high-dose topiramate treatment decreased LPP amplitudes in response to both emotional (pleasant and unpleasant) and drug-related cues (alcohol and cigarette), but not to neutral cues. However, results showed that the LPPs were not significant mediators of the relationship between topiramate dose and post-quit measures of substance use, reinforcement, craving, or withdrawal. CONCLUSIONS These findings suggest that high-dose topiramate (up to 250 mg/day) decreases the motivational salience of both drug-related and emotional cues among smokers with AUD. However, the nonsignificant mediation analyses preclude any firm conclusions about whether this effect represents one of topiramate's therapeutic mechanisms of action.
Collapse
Affiliation(s)
- Jason D. Robinson
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yong Cui
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maher Karam-Hage
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - George Kypriotakis
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Francesco Versace
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nassima Ait-Daoud Tiouririne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Robert M. Anthenelli
- Pacific Treatment and Research Center, Department of Psychiatry, University of California, San Diego, Health Sciences, La Jolla, California, USA
| | - Paul M. Cinciripini
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
14
|
Ilari A, Curti L, Petrella M, Cannella N, La Rocca A, Ranieri G, Gerace E, Iezzi D, Silvestri L, Mannaioni G, Ciccocioppo R, Masi A. Moderate ethanol drinking is sufficient to alter Ventral Tegmental Area dopamine neurons activity via functional and structural remodeling of GABAergic transmission. Neuropharmacology 2022; 203:108883. [PMID: 34785165 DOI: 10.1016/j.neuropharm.2021.108883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
Earlier studies have shown a major involvement of Ventral Tegmental Area (VTA) dopamine (DA) neurons in mediating the rewarding effects of ethanol (EtOH). Much less is known on the role of this system in mediating the transition from moderate to excessive drinking and abuse. Here we sought to explore the hypothesis that early stage drinking in rodents, resembling recreational EtOH use in humans, is sufficient to dysregulate VTA DA transmission thus increasing the propensity to use over time. To this purpose, midbrain slice recordings in mice previously exposed to an escalating (3, 6 and 12%) 18-day voluntary EtOH drinking paradigm was used. By recording from DA and γ-aminobutyric acid (GABA) VTA neurons in midbrain slices, we found that moderate EtOH drinking leads to a significant suppression of the spontaneous activity of VTA DA neurons, while increasing their response to acute EtOH application. We also found that chronic EtOH leads to the enhancement of GABA input frequency onto a subset of DA neurons. Structurally, chronic EtOH induced a significant increase in the number of GABA axonal boutons contacting DA neurons, suggesting deep rewiring of the GABA network. This scenario is consistent with a downmodulation of the reward DA system induced by moderate EtOH drinking, a neurochemical state defined as "hypodopaminergic" and previously associated with advanced stages of drug use in humans. In this context, increased sensitivity of DA neurons towards acute EtOH may represent the neurophysiological correlate of increased unitary rewarding value, possibly driving progression to addiction.
Collapse
Affiliation(s)
- A Ilari
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - L Curti
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - M Petrella
- Scuola di Scienze del Farmaco e dei Prodotti della salute, Università di Camerino, Italy
| | - N Cannella
- Scuola di Scienze del Farmaco e dei Prodotti della salute, Università di Camerino, Italy
| | - A La Rocca
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - G Ranieri
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - E Gerace
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - D Iezzi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - L Silvestri
- Dipartimento di Fisica ed Astronomia, Università di Firenze, Italy; European Laboratory for Non-linear Spectroscopy, Italy
| | - G Mannaioni
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy
| | - R Ciccocioppo
- Scuola di Scienze del Farmaco e dei Prodotti della salute, Università di Camerino, Italy.
| | - A Masi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, NEUROFARBA, Università di Firenze, Italy.
| |
Collapse
|
15
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
16
|
Althobaiti YS, Almutairi FM, Alshehri FS, Altowairqi E, Marghalani AM, Alghorabi AA, Alsanie WF, Gaber A, Alsaab HO, Almalki AH, Hakami AY, Alkhalifa T, Almalki AD, Hardy AMG, Shah ZA. Involvement of the dopaminergic system in the reward-related behavior of pregabalin. Sci Rep 2021; 11:10577. [PMID: 34011976 PMCID: PMC8134490 DOI: 10.1038/s41598-021-88429-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/08/2021] [Indexed: 01/23/2023] Open
Abstract
There has been an increase in cases of drug addiction and prescription drug abuse worldwide. Recently, pregabalin abuse has been a focus for many healthcare agencies, as highlighted by epidemiological studies. We previously evaluated the possibility of pregabalin abuse using the conditioned place preference (CPP) paradigm. We observed that a 60 mg/kg dose could induce CPP in mice and that pregabalin-rewarding properties were mediated through glutamate neurotransmission. Notably, the dopaminergic reward circuitry is also known to play a crucial role in medication-seeking behavior. Therefore, this study aimed to explore the possible involvement of dopaminergic receptor-1 in pregabalin-induced CPP. Mice were randomly allocated to receive saline or the dopamine-1 receptor antagonist SKF-83566 (0.03 mg/kg, intraperitoneal). After 30 min, the mice received either saline or pregabalin (60 mg/kg) during the conditioning phase. Among the control groups that received saline or SKF-83566, the time spent in the two conditioning chambers was not significantly altered. However, among the pregabalin-treated group, there was a marked increase in the time spent in the drug-paired chamber compared to the time spent in the vehicle-paired chamber. Notably, blocking dopamine-1 receptors with SKF-83566 completely prevented pregabalin-induced place preference, thus demonstrating the engagement of the dopaminergic system in pregabalin-induced reward-related behavior.
Collapse
Affiliation(s)
- Yusuf S Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia.
| | - Farooq M Almutairi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Deanship of Scientific Research, Taif University, Taif, 21944, Saudi Arabia
- Department of Clinical Laboratories Sciences, University of Hafar Al-Batin, College of Clinical Laboratories Sciences, Hafar Al-Batin, 39923, Saudi Arabia
| | - Fahad S Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Ebtehal Altowairqi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Aliyah M Marghalani
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Amal A Alghorabi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Walaa F Alsanie
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Ahmed Gaber
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Department of Biology, Faculty of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Hashem O Alsaab
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, 21944, Saudi Arabia
| | - Atiah H Almalki
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Alqassem Y Hakami
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Turki Alkhalifa
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia
| | - Ahmad D Almalki
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia
| | - Ana M G Hardy
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
17
|
Nunes-Freitas AL, Soni N, Polli FS, Kohlmeier KA. Prenatal exposure to nicotine in mice is associated with alterations in development and cellular and synaptic effects of alcohol in a brainstem arousal nucleus. Neurotoxicol Teratol 2021; 87:106980. [PMID: 33838245 DOI: 10.1016/j.ntt.2021.106980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023]
Abstract
Using drugs of abuse while pregnant has tremendous negative consequences for the offspring, including an enhanced risk for substance use disorder (SUD). This vulnerability suggests that gestational exposure to drugs alters the developmental trajectory of neurons important in SUD processes, which could lead to later life changes in responsiveness to motivationally salient stimuli. The laterodorsal tegmentum (LDT) gates the behaviorally relevant firing pattern signaling stimuli saliency in mesoaccumbal circuits. Accordingly, any alterations in LDT functionality could alter output, and play a role in negative outcomes on motivated behavior associated with early-life nicotine exposure. Therefore, we investigated whether prenatal exposure to nicotine (PNE), which is a known teratogen, altered responsiveness of LDT neurons to alcohol by conducting electrophysiology in brain slices. Alcohol induced an outward current in control LDT cells, which was not seen in PNE LDT neurons. The frequency of mEPSCs was significantly decreased by alcohol in LDT PNE cells and accompanied by a decrease in action potential frequency, which were actions not seen in controls. Changes in baseline activity of PNE LDT cells were also observed. In summary, PNE LDT neurons showed alterations in baseline activity and membrane and synaptic responses to postnatal exposures to alcohol. The differences in PNE baseline activity and alcohol responses likely lead to differential output from the LDT to mesoaccumbal targets that could play a role in biasing coding of relevant stimuli, which could participate in the enhanced proclivity for development of SUD in those exposed during gestation to nicotine.
Collapse
Affiliation(s)
- André Luiz Nunes-Freitas
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark; Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Neeraj Soni
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Filip S Polli
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristi A Kohlmeier
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
18
|
Driscoll JR, Wallace TL, Mansourian KA, Martin WJ, Margolis EB. Differential Modulation of Ventral Tegmental Area Circuits by the Nociceptin/Orphanin FQ System. eNeuro 2020; 7:ENEURO.0376-19.2020. [PMID: 32747458 PMCID: PMC7840174 DOI: 10.1523/eneuro.0376-19.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
The neuropeptide nociceptin/orphanin FQ (N/OFQ) can be released by stressors and is associated with disorders of emotion regulation and reward processing. N/OFQ and its receptor, NOP, are enriched in dopaminergic pathways, and intra-ventricular agonist delivery decreases dopamine levels in the dorsal striatum, nucleus accumbens (NAc), and ventral tegmental area (VTA). We used whole-cell electrophysiology in acute rat midbrain slices to investigate synaptic actions of N/OFQ. N/OFQ was primarily inhibitory, causing outward currents in both immunocytochemically identified dopaminergic (tyrosine hydroxylase positive (TH(+))) and non-dopaminergic (TH(-)) VTA neurons; effect at 1 μm: 20 ± 4 pA. Surprisingly, this effect was mediated by augmentation of postsynaptic GABAAR currents, unlike the substantia nigra pars compacta (SNc), where the N/OFQ-induced outward currents were K+ channel dependent. A smaller population, 17% of all VTA neurons, responded to low concentrations of N/OFQ with inward currents (10 nm: -11 ± 2 pA). Following 100 nm N/OFQ, the response to a second N/OFQ application was markedly diminished in VTA neurons (14 ± 10% of first response) but not in SNc neurons (90 ± 20% of first response). N/OFQ generated outward currents in medial prefrontal cortex (mPFC)-projecting VTA neurons, but inward currents in a subset of posterior anterior cingulate cortex (pACC)-projecting VTA neurons. While N/OFQ inhibited NAc-projecting VTA cell bodies, it had little effect on electrically or optogenetically evoked terminal dopamine release in the NAc measured ex vivo with fast scan cyclic voltammetry (FSCV). These results extend our understanding of the N/OFQ system in brainstem circuits implicated in many neurobehavioral disorders.
Collapse
Affiliation(s)
- Joseph R Driscoll
- BlackThorn Therapeutics, San Francisco, CA 94103
- UCSF Weill Institute of Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | | | - Kasra A Mansourian
- UCSF Weill Institute of Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | | | - Elyssa B Margolis
- UCSF Weill Institute of Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
19
|
Crummy EA, O'Neal TJ, Baskin BM, Ferguson SM. One Is Not Enough: Understanding and Modeling Polysubstance Use. Front Neurosci 2020; 14:569. [PMID: 32612502 PMCID: PMC7309369 DOI: 10.3389/fnins.2020.00569] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Substance use disorder (SUD) is a chronic, relapsing disease with a highly multifaceted pathology that includes (but is not limited to) sensitivity to drug-associated cues, negative affect, and motivation to maintain drug consumption. SUDs are highly prevalent, with 35 million people meeting criteria for SUD. While drug use and addiction are highly studied, most investigations of SUDs examine drug use in isolation, rather than in the more prevalent context of comorbid substance histories. Indeed, 11.3% of individuals diagnosed with a SUD have concurrent alcohol and illicit drug use disorders. Furthermore, having a SUD with one substance increases susceptibility to developing dependence on additional substances. For example, the increased risk of developing heroin dependence is twofold for alcohol misusers, threefold for cannabis users, 15-fold for cocaine users, and 40-fold for prescription misusers. Given the prevalence and risk associated with polysubstance use and current public health crises, examining these disorders through the lens of co-use is essential for translatability and improved treatment efficacy. The escalating economic and social costs and continued rise in drug use has spurred interest in developing preclinical models that effectively model this phenomenon. Here, we review the current state of the field in understanding the behavioral and neural circuitry in the context of co-use with common pairings of alcohol, nicotine, cannabis, and other addictive substances. Moreover, we outline key considerations when developing polysubstance models, including challenges to developing preclinical models to provide insights and improve treatment outcomes.
Collapse
Affiliation(s)
- Elizabeth A Crummy
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States.,Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Timothy J O'Neal
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States.,Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Britahny M Baskin
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States.,Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Susan M Ferguson
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Alcohol and Drug Abuse Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
20
|
Campos-Jurado Y, Martí-Prats L, Morón JA, Polache A, Granero L, Hipólito L. Dose-dependent induction of CPP or CPA by intra-pVTA ethanol: Role of mu opioid receptors and effects on NMDA receptors. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109875. [PMID: 31978422 PMCID: PMC7096259 DOI: 10.1016/j.pnpbp.2020.109875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 12/16/2022]
Abstract
The neurobiological mechanisms underlying alcohol motivational properties are still not fully understood, however, the mu-opioid receptors (MORs) have been evidenced as central elements in the manifestation of the alcohol reinforcing properties. Drug-associated environmental stimuli can trigger alcohol relapse and promote alcohol consumption whereby N-methyl-d-aspartate (NMDA) receptors play a pivotal role. Here we sought to demonstrate, for the first time, that ethanol induces conditioned place preference or aversion (CPP or CPA) when administered locally into the ventral tegmental area (VTA) and the associated role of MORs. We further analyzed the changes in the expression and mRNA levels of GluN1 and GluN2A subunits in designated brain areas. The expression of CPP or CPA was characterized following intra-VTA ethanol administration and we showed that either reinforcing (CPP) or aversive (CPA) properties are dependent on the dose administered (ranging here from 35 to 300 nmol). Furthermore, the critical contribution of local MORs in the acquisition of CPP was revealed by a selective antagonist, namely β-Funaltrexamine. Finally, modifications of the expression of NMDA receptor subunits in the Nucleus Accumbens (NAc) and Hippocampus after ethanol-induced CPP were analyzed at the proteomic and transcriptomic levels by western blot and In Situ Hybridation RNAscope techniques, respectively. Results showed that the mRNA levels of GluN2A but not GluN1 in NAc are higher after ethanol CPP. These novel results pave the way for further characterisation of the mechanisms by which ethanol motivational properties are associated with learned environmental cues.
Collapse
Affiliation(s)
- Yolanda Campos-Jurado
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Lucía Martí-Prats
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Jose A Morón
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Luis Granero
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain.
| |
Collapse
|
21
|
Cofresí RU, Bartholow BD, Piasecki TM. Evidence for incentive salience sensitization as a pathway to alcohol use disorder. Neurosci Biobehav Rev 2019; 107:897-926. [PMID: 31672617 PMCID: PMC6878895 DOI: 10.1016/j.neubiorev.2019.10.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
The incentive salience sensitization (ISS) theory of addiction holds that addictive behavior stems from the ability of drugs to progressively sensitize the brain circuitry that mediates attribution of incentive salience (IS) to reward-predictive cues and its behavioral manifestations. In this article, we establish the plausibility of ISS as an etiological pathway to alcohol use disorder (AUD). We provide a comprehensive and critical review of evidence for: (1) the ability of alcohol to sensitize the brain circuitry of IS attribution and expression; and (2) attribution of IS to alcohol-predictive cues and its sensitization in humans and non-human animals. We point out gaps in the literature and how these might be addressed. We also highlight how individuals with different alcohol subjective response phenotypes may differ in susceptibility to ISS as a pathway to AUD. Finally, we discuss important implications of this neuropsychological mechanism in AUD for psychological and pharmacological interventions attempting to attenuate alcohol craving and cue reactivity.
Collapse
Affiliation(s)
- Roberto U Cofresí
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States.
| | - Bruce D Bartholow
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| | - Thomas M Piasecki
- University of Missouri, Department of Psychological Sciences, Columbia, MO 65211, United States
| |
Collapse
|
22
|
Morel C, Montgomery S, Han MH. Nicotine and alcohol: the role of midbrain dopaminergic neurons in drug reinforcement. Eur J Neurosci 2019; 50:2180-2200. [PMID: 30251377 PMCID: PMC6431587 DOI: 10.1111/ejn.14160] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
Nicotine and alcohol addiction are leading causes of preventable death worldwide and continue to constitute a huge socio-economic burden. Both nicotine and alcohol perturb the brain's mesocorticolimbic system. Dopamine (DA) neurons projecting from the ventral tegmental area (VTA) to multiple downstream structures, including the nucleus accumbens, prefrontal cortex, and amygdala, are highly involved in the maintenance of healthy brain function. VTA DA neurons play a crucial role in associative learning and reinforcement. Nicotine and alcohol usurp these functions, promoting reinforcement of drug taking behaviors. In this review, we will first describe how nicotine and alcohol individually affect VTA DA neurons by examining how drug exposure alters the heterogeneous VTA microcircuit and network-wide projections. We will also examine how coadministration or previous exposure to nicotine or alcohol may augment the reinforcing effects of the other. Additionally, this review briefly summarizes the role of VTA DA neurons in nicotine, alcohol, and their synergistic effects in reinforcement and also addresses the remaining questions related to the circuit-function specificity of the dopaminergic system in mediating nicotine/alcohol reinforcement and comorbidity.
Collapse
Affiliation(s)
- Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Montgomery
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
23
|
Abstract
Ethanol produces intoxication through actions on numerous molecular and cellular targets. Adaptations involving these and other targets contribute to chronic drug actions that underlie continued and problematic drinking. Among the mechanisms involved in these ethanol actions are alterations in presynaptic mechanisms of synaptic transmission, including presynaptic protein function and excitation-secretion coupling. At synapses in the central nervous system (CNS), excitation-secretion coupling involves ion channel activation followed by vesicle fusion and neurotransmitter release. These mechanisms are altered by presynaptic neurotransmitter receptors and prominently by G protein-coupled receptors (GPCRs). Studies over the last 20-25 years have revealed that acute ethanol exposure alters neurotransmitter secretion, with especially robust effects on synapses that use the neurotransmitter gamma-aminobutyric acid (GABA). Intracellular signaling pathways involving second messengers such as cyclic AMP and calcium are implicated in these acute ethanol actions. Ethanol-induced release of neuropeptides and small molecule neurotransmitters that act on presynaptic GPCRs also contribute to presynaptic potentiation at synapses in the amygdala and hippocampus and inhibition of GABA release in the striatum. Prolonged exposure to ethanol alters neurotransmitter release at many CNS GABAergic and glutamatergic synapses, and changes in GPCR function are implicated in many of these neuroadaptations. These presynaptic neuroadaptations appear to involve compensation for acute drug effects at some synapses, but "allostatic" effects that result in long-term resetting of synaptic efficacy occur at others. Current investigations are determining how presynaptic neuroadaptations contribute to behavioral changes at different stages of alcohol drinking, with increasing focus on circuit adaptations underlying these behaviors. This chapter will discuss the acute and chronic presynaptic effects of ethanol in the CNS, as well as some of the consequences of these effects in amygdala and corticostriatal circuits that are related to excessive seeking/drinking and ethanol abuse.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
24
|
Bassareo V, Talani G, Frau R, Porru S, Rosas M, Kasture SB, Peana AT, Loi E, Sanna E, Acquas E. Inhibition of Morphine- and Ethanol-Mediated Stimulation of Mesolimbic Dopamine Neurons by Withania somnifera. Front Neurosci 2019; 13:545. [PMID: 31275092 PMCID: PMC6593272 DOI: 10.3389/fnins.2019.00545] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
Morphine- and ethanol-induced stimulation of neuronal firing of ventral tegmental area (VTA) dopaminergic neurons and of dopamine (DA) transmission in the shell of the nucleus accumbens (AcbSh) represents a crucial electrophysiological and neurochemical response underlying the ability of these compounds to elicit motivated behaviors and trigger a cascade of plasticity-related biochemical events. Previous studies indicate that the standardized methanolic extract of Withania somnifera roots (WSE) prevents morphine- and ethanol-elicited conditioned place preference and oral ethanol self-administration. Aim of the present research was to investigate whether WSE may also interfere with the ability of morphine and ethanol to stimulate VTA dopaminergic neurons and thus AcbSh DA transmission as assessed in male Sprague-Dawley rats by means of patch-clamp recordings in mesencephalic slices and in vivo brain microdialysis, respectively. Morphine and ethanol significantly stimulated spontaneous firing rate of VTA neurons and DA transmission in the AcbSh. WSE, at concentrations (200-400 μg/ml) that significantly reduce spontaneous neuronal firing of VTA DA neurons via a GABAA- but not GABAB-mediated mechanism, suppressed the stimulatory actions of both morphine and ethanol. Moreover, in vivo administration of WSE at a dose (75 mg/kg) that fails to affect basal DA transmission, significantly prevented both morphine- and ethanol-elicited increases of DA in the AcbSh. Overall, these results highlight the ability of WSE to interfere with morphine- and ethanol-mediated central effects and suggest a mechanistic interpretation of the efficacy of this extract to prevent the motivational properties of these compounds.
Collapse
Affiliation(s)
- Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,Centre of Excellence on Neurobiology of Addiction, University of Cagliari, Cagliari, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Simona Porru
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Michela Rosas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | | | - Alessandra T Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Eleonora Loi
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Enrico Sanna
- Centre of Excellence on Neurobiology of Addiction, University of Cagliari, Cagliari, Italy.,Institute of Neuroscience, National Research Council, Cagliari, Italy.,Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Elio Acquas
- Centre of Excellence on Neurobiology of Addiction, University of Cagliari, Cagliari, Italy.,Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
25
|
Ho SY, Chien YH, Tsai LK, Muramatsu SI, Hwu WL, Liou HH, Lee NC. Electrical Abnormalities in Dopaminergic Neurons of the Substantia Nigra in Mice With an Aromatic L-Amino Acid Decarboxylase Deficiency. Front Cell Neurosci 2019; 13:9. [PMID: 30766478 PMCID: PMC6365702 DOI: 10.3389/fncel.2019.00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/10/2019] [Indexed: 12/25/2022] Open
Abstract
Aromatic L-acid decarboxylase (AADC) deficiency causes severe motor disturbances in affected children. A putamen-targeted gene therapy improves the motor function of patients. The present study investigated the electrical properties of dopaminergic (DA) neurons in the substantia nigra compacta (SNc) of mice with an AADC deficiency (DdcKI). The basal firing of DA neurons, which determines DA release in the putamen, was abnormal in the DdcKI mice, including a low frequency and irregular firing pattern, because of a decrease in the after-hyperpolarization (AHP) amplitude of action potentials (APs). The frequency of spontaneous excitatory postsynaptic currents (sEPSCs) increased and that of spontaneous inhibitory PSCs (sIPSCs) decreased in the SNc DA neurons from the DdcKI mice, suggesting an elevation in glutamatergic excitatory stimuli and a reduction in GABAergic inhibitory stimuli, respectively. Altered expression patterns of genes encoding receptors and channels were also observed in the DdcKI mice. Administration of a widespread neuron-specific gene therapy to the brains of the DdcKI mice partially corrected these electric abnormalities. The overexcitability of SNc DA neurons in the presence of generalized dopamine deficiency likely underlies the occurrence of motor disturbances.
Collapse
Affiliation(s)
- Shih-Yin Ho
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shin-Ichi Muramatsu
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan.,Center for Gene & Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Horng-Huei Liou
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
26
|
Zhou C, Gu W, Wu H, Yan X, Deshpande P, Xiao C, Lester HA. Bidirectional dopamine modulation of excitatory and inhibitory synaptic inputs to subthalamic neuron subsets containing α4β2 or α7 nAChRs. Neuropharmacology 2019; 148:220-228. [PMID: 30660626 DOI: 10.1016/j.neuropharm.2019.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 02/05/2023]
Abstract
The subthalamic nucleus (STN) possesses microcircuits distinguished by subtypes of nicotinic acetylcholine receptors (nAChRs). Although dysfunction of the STN is well-known in Parkinson's disease, there is still little information about whether dopamine differentially modulates excitatory and inhibitory synaptic inputs to STN neurons expressing different nAChR subtypes. To address this issue, we performed brain slice patch-clamp recordings on STN neurons, while we pharmacologically manipulated dopaminergic inputs. In STN neuron subsets containing either α4β2 or α7 nAChRs, D1 and D2 receptors respectively enhanced and inhibited spontaneous inhibitory and excitatory postsynaptic currents (sIPSCs and sEPSCs) and firing rates. The elevation of dopamine levels resulted in diverse regulations of synaptic transmission in these two neuron subsets, and interestingly, the dopamine regulation of sIPSCs significantly correlated with that of sEPSCs. Surprisingly, depletion of dopamine either by reserpine treatment or by unilateral 6-OHDA lesion of nigrostriatal dopaminergic neurons did not alter synaptic inputs to STN neurons, but STN neurons in the 6-OHDA-lesioned side exhibited hyperactivity. In summary, dopamine regulated both GABAergic and glutamatergic synaptic inputs to STN neuron subsets containing either α4β2 or α7 nAChRs, forming a balancing machinery to control neuronal activity. In parkinsonian mice, postsynaptic mechanisms may exist and contribute to the hyperactivity of STN neurons.
Collapse
Affiliation(s)
- Chunyi Zhou
- Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Weixin Gu
- Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Haichuan Wu
- Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xiang Yan
- Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Purnima Deshpande
- Division of Biology, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Cheng Xiao
- Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; Division of Biology, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Henry A Lester
- Division of Biology, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
27
|
You C, Savarese A, Vandegrift BJ, He D, Pandey SC, Lasek AW, Brodie MS. Ethanol acts on KCNK13 potassium channels in the ventral tegmental area to increase firing rate and modulate binge-like drinking. Neuropharmacology 2019; 144:29-36. [PMID: 30332606 PMCID: PMC6286249 DOI: 10.1016/j.neuropharm.2018.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/27/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022]
Abstract
Alcohol excitation of the ventral tegmental area (VTA) is important in neurobiological processes related to the development of alcoholism. The ionotropic receptors on VTA neurons that mediate ethanol-induced excitation have not been identified. Quinidine blocks ethanol excitation of VTA neurons, and blockade of two-pore potassium channels is among the actions of quinidine. Therefore two-pore potassium channels in the VTA may be potential targets for the action of ethanol. Here, we explored whether ethanol activation of VTA neurons is mediated by the two-pore potassium channel KCNK13. Extracellular recordings of the response of VTA neurons to ethanol were performed in combination with knockdown of Kcnk13 using a short hairpin RNA (shRNA) in C57BL/6 J mice. Real-time PCR and immunohistochemistry were used to examine expression of this channel in the VTA. Finally, the role of KCNK13 in binge-like drinking was examined in the drinking in the dark test after knockdown of the channel. Kcnk13 expression in the VTA was increased by acute ethanol exposure. Ethanol-induced excitation of VTA neurons was selectively reduced by shRNA targeting Kcnk13. Importantly, knockdown of Kcnk13 in the VTA resulted in increased alcohol drinking. These results are consistent with the idea that ethanol stimulates VTA neurons at least in part by inhibiting KCNK13, a specific two-pore potassium channel, and that KCNK13 can control both VTA neuronal activity and binge drinking. KCNK13 is a novel alcohol-sensitive molecular target and may be amenable to the development of pharmacotherapies for alcoholism treatment.
Collapse
Affiliation(s)
- Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Antonia Savarese
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bertha J Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Donghong He
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
28
|
Extension of Reward-Attention Circuit Model: Alcohol’s Influence on Attentional Focus and Consequences on Autism Spectrum Disorder. Neurocomputing 2019. [DOI: 10.1016/j.neucom.2018.10.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
29
|
Jose NA, Yadav P, Kapoor A, Mahla VP. Comparison between baclofen and topiramate in alcohol dependence: A prospective study. Ind Psychiatry J 2019; 28:44-50. [PMID: 31879446 PMCID: PMC6929233 DOI: 10.4103/ipj.ipj_57_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/19/2019] [Accepted: 09/24/2019] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Baclofen and topiramate are GABAergic drugs, and both have been recommended for the treatment of alcohol dependence as anticraving agent. Several studies have demonstrated the effect of baclofen and topiramate as anticraving, but none has compared them. The main aim of the current study was to assess the baclofen and topiramate as anticraving agent in alcohol dependence during 1 month follow-up. METHODOLOGY After 1-week detoxification protocol, 94 patients were randomly assigned to either baclofen (n = 49) or topiramate (n = 45) for 1-month follow-up. Patients were assessed with clinical institute withdrawal assessment at baseline, and at 1 week, the Addiction Severity Index, ready to change questionnaire at baseline and weekly assessed with Obsessive and Compulsive drinking scale (OCDS) for craving. At every follow-up, adverse effects were also assessed to check tolerability. RESULTS A marked improvement was observed with baclofen in OCDS in each assessment as compared to topiramate. With baclofen, 61.22% of patients became complete abstinence, as compared to 37.78% in topiramate group. Baclofen was better tolerated as 24.49% patients were dropped out in baclofen group as compared to 33.33% in topiramate group. CONCLUSION Baclofen has better efficacy and tolerability as compared to topiramate.
Collapse
Affiliation(s)
- Nimmi A Jose
- Department of Psychiatry, Shri Guru Gobind Tricentenary Medical College and Hospital, Gurugram, Haryana, India
| | - Pooja Yadav
- Department of Psychiatry, Shri Guru Gobind Tricentenary Medical College and Hospital, Gurugram, Haryana, India
| | - Abhishek Kapoor
- Department of Psychiatry, Shri Guru Gobind Tricentenary Medical College and Hospital, Gurugram, Haryana, India
| | - Ved Pal Mahla
- Department of Psychiatry, Shri Guru Gobind Tricentenary Medical College and Hospital, Gurugram, Haryana, India
| |
Collapse
|
30
|
Wang T, Zhu H, Hou Y, Gu W, Wu H, Luan Y, Xiao C, Zhou C. Galantamine reversed early postoperative cognitive deficit via alleviating inflammation and enhancing synaptic transmission in mouse hippocampus. Eur J Pharmacol 2018; 846:63-72. [PMID: 30586550 DOI: 10.1016/j.ejphar.2018.12.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/20/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is commonly seen in patients undergoing major surgeries and may persist. Although neuroinflammation is one of the important contributors to the development of POCD, the mechanisms underlying POCD remain unclear. We performed stabilized tibial fracture operation in male mice. In comparison with sham mice (anesthesia only), the surgery mice exhibited cognitive deficits in a fear conditioning paradigm at postsurgery day 3-7, and increased numbers of microglia and elevated levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) without change of anti-inflammatory cytokines (IL-4 and IL-10) in the hippocampus. Electrophysiological recordings from CA1 hippocampal neurons revealed that POCD mice exhibited impairment in AMPA receptor-mediated evoked excitatory postsynaptic currents (eEPSCs) without alteration in the rectification property of AMPA receptors. Interestingly, daily intraperitoneal administration of galantamine, an inhibitor of acetylcholinesterase, reversed cognitive dysfunction in surgery mice and attenuated accumulation of microglia and protein levels of IL-1β, IL-6 and TNF-α in the hippocampus. Additionally, galantamine potentiated AMPA receptor-mediated eEPSCs in the hippocampus more prominent in surgery mice than in sham mice. Therefore, enhancement of cholinergic tone in the hippocampus might be a therapeutic strategy for early POCD in terms of suppression of inflammation and normalization of excitatory synaptic transmission.
Collapse
Affiliation(s)
- Tianhai Wang
- Department of Anesthesiology, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hongge Zhu
- Department of Second Pulmonary Medicine, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yanshen Hou
- Department of Anesthesiology, The third hospital, affiliated to the Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Weixin Gu
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haichuan Wu
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yiwen Luan
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cheng Xiao
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chunyi Zhou
- Jiangsu Province Key laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
31
|
Shillinglaw JE, Morrisett RA, Mangieri RA. Ethanol Modulates Glutamatergic Transmission and NMDAR-Mediated Synaptic Plasticity in the Agranular Insular Cortex. Front Pharmacol 2018; 9:1458. [PMID: 30618752 PMCID: PMC6305468 DOI: 10.3389/fphar.2018.01458] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022] Open
Abstract
The agranular insular cortex (AIC) has recently been investigated by the alcohol field because of its connectivity to and modulatory control over limbic and brainstem regions implicated in alcohol use disorder (AUD), and because it has shown involvement in animal models of alcohol drinking. Despite evidence of AIC involvement in AUD, there has not yet been an examination of whether ethanol modulates glutamatergic and γ-amino-butyric acid (GABA)ergic synaptic transmission and plasticity in the AIC. Characterizing how the synaptic transmission and plasticity states of AIC cortical processing neurons are modulated by acute ethanol will likely reveal the molecular targets by which chronic ethanol alters AIC function as alcohol drinking transitions from controlled to problematic. Therefore, we collected brain slices from ethanol-naïve adult male mice, obtained whole-cell recording configuration in layer 2/3 AIC pyramidal neurons, and bath-applied ethanol at pharmacologically relevant concentrations during electrophysiological assays of glutamatergic and GABAergic synaptic transmission and plasticity. We found that ethanol inhibited electrically evoked N-methyl-D-aspartate receptor (NMDAR)-mediated excitatory post-synaptic currents (EPSCs) in a concentration-related fashion, and had little effect on evoked α-amino-3-hydrox-5-methylisoxazole-4-propionic acid-type receptor (AMPAR)-mediated EPSCs. Ethanol had no effect on spontaneous excitatory post-synaptic currents (sEPSCs) or inhibitory GABAAR-mediated post-synaptic currents (sIPSCs). We found that synaptic conditioning (low-frequency stimulation for 15 min at 1 Hz) induced a form of long-term depression (LTD) of evoked AMPAR-mediated EPSCs. The ability to induce LTD was inhibited by a non-selective NMDAR antagonist (DL-2-amino-5-phosphonovaleric acid), and also by acute, intoxicating concentrations of ethanol. Taken together these data suggest that the glutamate, but not GABA system in the AIC is uniquely sensitive to ethanol, and that in particular NMDAR-mediated processes in the AIC may be disrupted by pharmacologically relevant concentrations of ethanol.
Collapse
Affiliation(s)
- Joel E Shillinglaw
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Richard A Morrisett
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Regina A Mangieri
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
32
|
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: 6.4] [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
|
33
|
di Volo M, Morozova EO, Lapish CC, Kuznetsov A, Gutkin B. Dynamical ventral tegmental area circuit mechanisms of alcohol-dependent dopamine release. Eur J Neurosci 2018; 50:2282-2296. [PMID: 30215874 DOI: 10.1111/ejn.14147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/15/2018] [Accepted: 08/24/2018] [Indexed: 11/28/2022]
Abstract
A large body of data has identified numerous molecular targets through which ethanol (EtOH) acts on brain circuits. Yet how these multiple mechanisms interact to result in dysregulated dopamine (DA) release under the influence of alcohol in vivo remains unclear. In this manuscript, we delineate potential circuit-level mechanisms responsible for EtOH-dependent dysregulation of DA release from the ventral tegmental area (VTA) into its projection areas. For this purpose, we constructed a circuit model of the VTA that integrates realistic Glutamatergic (Glu) inputs and reproduces DA release observed experimentally. We modelled the concentration-dependent effects of EtOH on its principal VTA targets. We calibrated the model to reproduce the inverted U-shape dose dependence of DA neuron activity on EtOH concentration. The model suggests a primary role of EtOH-induced boost in the Ih and AMPA currents in the DA firing-rate/bursting increase. This is counteracted by potentiated GABA transmission that decreases DA neuron activity at higher EtOH concentrations. Thus, the model connects well-established in vitro pharmacological EtOH targets with its in vivo influence on neuronal activity. Furthermore, we predict that increases in VTA activity produced by moderate EtOH doses require partial synchrony and relatively low rates of the Glu afferents. We propose that the increased frequency of transient (phasic) DA peaks evoked by EtOH results from synchronous population bursts in VTA DA neurons. Our model predicts that the impact of acute ETOH on dopamine release is critically shaped by the structure of the cortical inputs to the VTA.
Collapse
Affiliation(s)
- Matteo di Volo
- Unité de Neurosciences, Information et Complexité, CNRS, Gif-sur-Yvette, France.,Group for Neural Theory, LNC INSERM U960, DEC Ecole Normale Superieure PSL University, Paris, France
| | | | - Christopher C Lapish
- Addiction Neuroscience Program, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Alexey Kuznetsov
- Department of Mathematical Sciences, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Boris Gutkin
- Group for Neural Theory, LNC INSERM U960, DEC Ecole Normale Superieure PSL University, Paris, France.,Center for Cognition and Decision Making, NRU HSE, Moscow, Russia
| |
Collapse
|
34
|
You C, Vandegrift B, Brodie MS. Ethanol actions on the ventral tegmental area: novel potential targets on reward pathway neurons. Psychopharmacology (Berl) 2018; 235:1711-1726. [PMID: 29549390 PMCID: PMC5949141 DOI: 10.1007/s00213-018-4875-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
Abstract
The ventral tegmental area (VTA) evaluates salience of environmental stimuli and provides dopaminergic innervation to many brain areas affected by acute and chronic ethanol exposure. While primarily associated with rewarding and reinforcing stimuli, recent evidence indicates a role for the VTA in aversion as well. Ethanol actions in the VTA may trigger neuroadaptation resulting in reduction of the aversive responses to alcohol and a relative increase in the rewarding responses. In searching for effective pharmacotherapies for the treatment of alcohol abuse and alcoholism, recognition of this imbalance may reveal novel strategies. In addition to conventional receptor/ion channel pharmacotherapies, epigenetic factors that control neuroadaptation to chronic ethanol treatment can be targeted as an avenue for development of therapeutic approaches to restore the balance. Furthermore, when exploring therapies to address reward/aversion imbalance in the action of alcohol in the VTA, sex differences have to be taken into account to ensure effective treatment for both men and women. These principles apply to a VTA-centric approach to therapies, but should hold true when thinking about the overall approach in the development of neuroactive drugs to treat alcohol use disorders. Although the functions of the VTA itself are complex, it is a useful model system to evaluate the reward/aversion imbalance that occurs with ethanol exposure and could be used to provide new leads in the efforts to develop novel drugs to treat alcoholism.
Collapse
Affiliation(s)
- Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bertha Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA.
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
35
|
Xiong M, Shiwalkar N, Reddy K, Shin P, Bekker A. Neurobiology of Propofol Addiction and Supportive Evidence: What Is the New Development? Brain Sci 2018; 8:brainsci8020036. [PMID: 29470436 PMCID: PMC5836055 DOI: 10.3390/brainsci8020036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/14/2018] [Accepted: 02/18/2018] [Indexed: 12/21/2022] Open
Abstract
Propofol is a short-acting intravenous anesthetic agent suitable for induction and maintenance of general anesthesia as well as for procedural and intensive care unit sedation. As such it has become an unparalleled anesthetic agent of choice in many institutional and office practices. However, in addition to its idealistic properties as an anesthetic agent, there is accumulating evidence suggesting its potential for abuse. Clinical and experimental evidence has revealed that not only does propofol have the potential to be abused, but also that addiction to propofol shows a high mortality rate. Based on this evidence, different researchers have shown interest in determining the probability of propofol to be an addictive agent by comparing it with other drugs of abuse and depicting a functional similitude that involves the mesocorticolimbic pathway of addiction. In light of this, the Drug Enforcement Agency and the American Society of Anesthesiologists have put forth certain safety recommendations for the use of propofol. Despite this, the abuse potential of propofol has been challenged at different levels and therefore the preeminent focus will be to further validate the linkage from medicinal and occasional use of propofol to its addiction, as well as to explore the cellular and molecular targets involved in establishing this linkage, so as to curb the harm arising out of it. This review incorporates the clinical and biomolecular evidence supporting the abuse potential of propofol and brings forth the promising targets and the foreseeable mechanism causing the propofol addiction phenotypes, which can be called upon for future developments in this field.
Collapse
Affiliation(s)
- Ming Xiong
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Nimisha Shiwalkar
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Kavya Reddy
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Peter Shin
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| |
Collapse
|
36
|
The Cerebellar GABA AR System as a Potential Target for Treating Alcohol Use Disorder. Handb Exp Pharmacol 2018; 248:113-156. [PMID: 29736774 DOI: 10.1007/164_2018_109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the brain, fast inhibitory neurotransmission is mediated primarily by the ionotropic subtype of the gamma-aminobutyric acid (GABA) receptor subtype A (GABAAR). It is well established that the brain's GABAAR system mediates many aspects of neurobehavioral responses to alcohol (ethanol; EtOH). Accordingly, in both preclinical studies and some clinical scenarios, pharmacologically targeting the GABAAR system can alter neurobehavioral responses to acute and chronic EtOH consumption. However, many of the well-established interactions of EtOH and the GABAAR system have been identified at concentrations of EtOH ([EtOH]) that would only occur during abusive consumption of EtOH (≥40 mM), and there are still inadequate treatment options for prevention of or recovery from alcohol use disorder (AUD, including abuse and dependence). Accordingly, there is a general acknowledgement that more research is needed to identify and characterize: (1) neurobehavioral targets of lower [EtOH] and (2) associated brain structures that would involve such targets in a manner that may influence the development and maintenance of AUDs.Nearly 15 years ago it was discovered that the GABAAR system of the cerebellum is highly sensitive to EtOH, responding to concentrations as low as 10 mM (as would occur in the blood of a typical adult human after consuming 1-2 standard units of EtOH). This high sensitivity to EtOH, which likely mediates the well-known motor impairing effects of EtOH, combined with recent advances in our understanding of the role of the cerebellum in non-motor, cognitive/emotive/reward processes has renewed interest in this system in the specific context of AUD. In this chapter we will describe recent advances in our understanding of cerebellar processing, actions of EtOH on the cerebellar GABAAR system, and the potential relationship of such actions to the development of AUD. We will finish with speculation about how cerebellar specific GABAAR ligands might be effective pharmacological agents for treating aspects of AUD.
Collapse
|
37
|
Abrahao KP, Salinas AG, Lovinger DM. Alcohol and the Brain: Neuronal Molecular Targets, Synapses, and Circuits. Neuron 2017; 96:1223-1238. [PMID: 29268093 PMCID: PMC6566861 DOI: 10.1016/j.neuron.2017.10.032] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/30/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022]
Abstract
Ethanol is one of the most commonly abused drugs. Although environmental and genetic factors contribute to the etiology of alcohol use disorders, it is ethanol's actions in the brain that explain (1) acute ethanol-related behavioral changes, such as stimulant followed by depressant effects, and (2) chronic changes in behavior, including escalated use, tolerance, compulsive seeking, and dependence. Our knowledge of ethanol use and abuse thus relies on understanding its effects on the brain. Scientists have employed both bottom-up and top-down approaches, building from molecular targets to behavioral analyses and vice versa, respectively. This review highlights current progress in the field, focusing on recent and emerging molecular, cellular, and circuit effects of the drug that impact ethanol-related behaviors. The focus of the field is now on pinpointing which molecular effects in specific neurons within a brain region contribute to behavioral changes across the course of acute and chronic ethanol exposure.
Collapse
Affiliation(s)
- Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Armando G Salinas
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Ostroumov A, Dani JA. Convergent Neuronal Plasticity and Metaplasticity Mechanisms of Stress, Nicotine, and Alcohol. Annu Rev Pharmacol Toxicol 2017; 58:547-566. [PMID: 28977763 DOI: 10.1146/annurev-pharmtox-010617-052735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress and tobacco smoking are risk factors for alcoholism, but the underlying neural mechanisms are not well understood. Although stress, nicotine, and alcohol have broad, individual effects in the brain, some of their actions converge onto the same mechanisms and circuits. Stress and nicotine augment alcohol-related behaviors, in part via modulation of alcohol-evoked neuronal plasticity and metaplasticity mechanisms. Stress modulates alcohol-evoked plasticity via the release of signaling molecules that influence synaptic transmission. Nicotine also activates some of the same signaling molecules, cells, and circuits, producing a convergence of both stress and nicotine onto common plasticity mechanisms that influence alcohol self-administration. We describe several forms of alcohol-induced plasticity, including classic Hebbian plasticity at glutamatergic synapses, and we highlight less appreciated forms, such as non-Hebbian and GABAergic synaptic plasticity. Risk factors such as stress and nicotine initiate lasting neural changes that modify subsequent alcohol-induced synaptic plasticity and increase the vulnerability to alcohol addiction.
Collapse
Affiliation(s)
- Alexey Ostroumov
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| |
Collapse
|
39
|
Shah A, Zuo W, Kang S, Li J, Fu R, Zhang H, Bekker A, Ye JH. The lateral habenula and alcohol: Role of glutamate and M-type potassium channels. Pharmacol Biochem Behav 2017. [PMID: 28624587 DOI: 10.1016/j.pbb.2017.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) or alcoholism is a chronic relapsing disorder. Our knowledge of alcoholism hinges on our understanding of its effects on the brain. This review will center on the effects of alcohol in the lateral habenula (LHb), an epithalamic structure that connects the forebrain with the midbrain and encodes aversive signaling. Like many addictive drugs, alcohol has both rewarding and aversive properties. While alcohol's euphoric property is believed to be important for the initiation of drinking, increasing evidence suggests that alcohol's negative affect plays a critical role in excessive drinking and alcohol dependence. During withdrawal and abstinence, alcoholics often experience anxiety and depressions, both of which have been implicated in relapse drinking. This review focuses on the recent accumulation of knowledge about the effects of acute and chronic alcohol exposure on the activity of and synaptic transmissions on LHb neurons, as well as the effects of manipulation of LHb function on alcohol consumption and related behaviors. Recent evidence highlights a critical role for the LHb in AUD and related psychiatric ailments. Multidisciplinary work in animals collectively suggests that LHb function and activity, including M-type potassium channels and glutamatergic transmission are altered by acute and repeated chronic alcohol exposure. We will also discuss how functional, pharmacological, and chemogenetic manipulation of the LHb affects ethanol drinking and psychiatric disorders occurring in animals withdrawn from chronic alcohol exposure. Conceivable mechanisms behind these effects and their potential as targets for therapies will also be discussed.
Collapse
Affiliation(s)
- Avi Shah
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Wanhong Zuo
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Seungwoo Kang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Jing Li
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Rao Fu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Haifeng Zhang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA.
| |
Collapse
|
40
|
Huang CCY, Ma T, Roltsch Hellard EA, Wang X, Selvamani A, Lu J, Sohrabji F, Wang J. Stroke triggers nigrostriatal plasticity and increases alcohol consumption in rats. Sci Rep 2017; 7:2501. [PMID: 28566754 PMCID: PMC5451385 DOI: 10.1038/s41598-017-02714-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Excessive alcohol consumption is a known risk factor for stroke, but the effect of stroke on alcohol intake is unknown. The dorsomedial striatum (DMS) and midbrain areas of the nigrostriatal circuit are critically associated to stroke and alcohol addiction. Here we sought to explore the influence of stroke on alcohol consumption and to uncover the underlying nigrostriatal mechanism. Rats were trained to consume alcohol using a two-bottle choice or operant self-administration procedure. Retrograde beads were infused into the DMS or midbrain to label specific neuronal types, and ischemic stroke was induced in the dorsolateral striatum (DLS). Slice electrophysiology was employed to measure excitability and synaptic transmission in DMS and midbrain neurons. We found that ischemic stroke-induced DLS infarction produced significant increases in alcohol preference, operant self-administration, and relapse. These increases were accompanied by enhanced excitability of DMS and midbrain neurons. In addition, glutamatergic inputs onto DMS D1-neurons was potentiated, whereas GABAergic inputs onto DMS-projecting midbrain dopaminergic neurons was suppressed. Importantly, systemic inhibition of dopamine D1 receptors attenuated the stroke-induced increase in operant alcohol self-administration. Our results suggest that the stroke-induced DLS infarction evoked abnormal plasticity in nigrostriatal dopaminergic neurons and DMS D1-neurons, contributing to increased post-stroke alcohol-seeking and relapse.
Collapse
Affiliation(s)
- Cathy C Y Huang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Tengfei Ma
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Emily A Roltsch Hellard
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | | | - Amutha Selvamani
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jiayi Lu
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
| |
Collapse
|
41
|
Harrison NL, Skelly MJ, Grosserode EK, Lowes DC, Zeric T, Phister S, Salling MC. Effects of acute alcohol on excitability in the CNS. Neuropharmacology 2017; 122:36-45. [PMID: 28479395 DOI: 10.1016/j.neuropharm.2017.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/23/2023]
Abstract
Alcohol has many effects on brain function and hence on human behavior, ranging from anxiolytic and mild disinhibitory effects, sedation and motor incoordination, amnesia, emesis, hypnosis and eventually unconsciousness. In recent years a variety of studies have shown that acute and chronic exposure to alcohol can modulate ion channels that regulate excitability. Modulation of intrinsic excitability provides another way in which alcohol can influence neuronal network activity, in addition to its actions on synaptic inputs. In this review, we review "low dose" effects [between 2 and 20 mM EtOH], and "medium dose"; effects [between 20 and 50 mM], by considering in turn each of the many networks and brain regions affected by alcohol, and thereby attempt to integrate in vitro physiological studies in specific brain regions (e.g. amygdala, ventral tegmental area, prefrontal cortex, thalamus, cerebellum etc.) within the context of alcohol's behavioral actions in vivo (e.g. anxiolysis, euphoria, sedation, motor incoordination). This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Neil L Harrison
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States.
| | - Mary Jane Skelly
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Emma K Grosserode
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Daniel C Lowes
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Tamara Zeric
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Sara Phister
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| | - Michael C Salling
- Departments of Anesthesiology and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, N.Y., 10032, United States
| |
Collapse
|
42
|
Zuo W, Fu R, Hopf FW, Xie G, Krnjević K, Li J, Ye JH. Ethanol drives aversive conditioning through dopamine 1 receptor and glutamate receptor-mediated activation of lateral habenula neurons. Addict Biol 2017; 22:103-116. [PMID: 26283508 DOI: 10.1111/adb.12298] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/15/2015] [Accepted: 07/23/2015] [Indexed: 12/11/2022]
Abstract
There has been increasing interest in the lateral habenula (LHb) given its potent regulatory role in many aversion-related behaviors. Interestingly, ethanol can be rewarding as well as aversive; we therefore investigated whether ethanol exposure alters pacemaker firing or glutamate receptor signaling in LHb neurons in vitro and also whether LHb activity in vivo might contribute to the acquisition of conditioned place aversion to ethanol. Surprisingly, in epithalamic slices, low doses of ethanol (1.4 mM) strongly accelerated LHb neuron firing (by ~60%), and ethanol's effects were much reduced by blocking glutamate receptors. Ethanol increased presynaptic glutamate release, and about half of this effect was mediated by dopamine subtype 1 receptors (D1Rs) and cyclic adenosine monophosphate (cAMP)-dependent signaling pathways. In agreement with these findings, c-Fos immunoreactivity in LHb regions was enhanced after a single administration of a low dose of ethanol (0.25 g/kg i.p.). Importantly, the same dose of ethanol in vivo also produced strong conditioned place aversion, and this was prevented by inhibiting D1Rs or neuronal activity within the LHb. By contrast, a higher dose (2 g/kg) led to ethanol conditioned place preference, which was enhanced by inhibiting neuronal activity or D1Rs within the LHb and suppressed by infusing aminomethylphosphonic acid or the D1R agonist SKF38393 within the LHb. Our in vitro and in vivo observations show, for the first time, that ethanol increases LHb excitation, mediated by D1R and glutamate receptors, and may underlie a LHb aversive signal that contributes to ethanol-related aversion.
Collapse
Affiliation(s)
- Wanhong Zuo
- Department of Anesthesiology, Pharmacology and Physiology, New Jersey Medical School; Rutgers, The State University of New Jersey; Newark NJ USA
| | - Rao Fu
- Department of Anesthesiology, Pharmacology and Physiology, New Jersey Medical School; Rutgers, The State University of New Jersey; Newark NJ USA
| | - Frederic Woodward Hopf
- Department of Neurology; University of California at San Francisco; San Francisco CA USA
| | - Guiqin Xie
- Department of Anesthesiology, Pharmacology and Physiology, New Jersey Medical School; Rutgers, The State University of New Jersey; Newark NJ USA
| | | | - Jing Li
- Department of Anesthesiology, Pharmacology and Physiology, New Jersey Medical School; Rutgers, The State University of New Jersey; Newark NJ USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology and Physiology, New Jersey Medical School; Rutgers, The State University of New Jersey; Newark NJ USA
| |
Collapse
|
43
|
Nimitvilai S, You C, Arora DS, McElvain MA, Vandegrift BJ, Brodie MS, Woodward JJ. Differential Effects of Toluene and Ethanol on Dopaminergic Neurons of the Ventral Tegmental Area. Front Neurosci 2016; 10:434. [PMID: 27713687 PMCID: PMC5031606 DOI: 10.3389/fnins.2016.00434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/07/2016] [Indexed: 11/13/2022] Open
Abstract
Drugs of abuse increase the activity of dopaminergic neurons of the ventral tegmental area (VTA), and output from the VTA is critical for both natural and drug-induced reward and reinforcement. Ethanol and the abused inhalant toluene both enhance VTA neuronal firing, but the mechanisms of this effect is not fully known. In this study, we used extracellular recordings to compare the actions of toluene and ethanol on DA VTA neurons. Both ethanol and toluene increased the firing rate of DA neurons, although toluene was ~100 times more potent than ethanol. The mixed ion channel blocker quinine (100 μM) blocked the increases in firing produced by ethanol and toluene, indicating some similarity in mechanisms of excitation. A mixture of antagonists of GABA and cholinergic receptors did not prevent toluene-induced or ethanol-induced excitation, and toluene-induced excitation was not altered by co-administration of ethanol, suggesting independent mechanisms of excitation for ethanol and toluene. Concurrent blockade of NMDA, AMPA, and metabotropic glutamate receptors enhanced the excitatory effect of toluene while having no significant effect on ethanol excitation. Nicotine increased firing of DA VTA neurons, and this was blocked by the nicotinic antagonist mecamylamine (1 μM). Mecamylamine did not alter ethanol or toluene excitation of firing but the muscarinic antagonist atropine (5 μM) or a combination of GABA antagonists (bicuculline and CGP35348, 10 μM each) reduced toluene-induced excitation without affecting ethanol excitation. The Ih current blocker ZD7288 abolished the excitatory effect of toluene but unlike the block of ethanol excitation, the effect of ZD7288 was not reversed by the GIRK channel blocker barium, but was reversed by GABA antagonists. These results demonstrate that the excitatory effects of ethanol and toluene have some similarity, such as block by quinine and ZD7288, but also indicate that there are important differences between these two drugs in their modulation by glutamatergic, cholinergic, and GABAergic receptors. These findings provide important information regarding the actions of abused inhalants on central reward pathways, and suggest that regulation of the activation of central dopamine pathways by ethanol and toluene partially overlap.
Collapse
Affiliation(s)
- Sudarat Nimitvilai
- Department of Neuroscience, Medical University of South Carolina Charleston, SC, USA
| | - Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago Chicago, IL, USA
| | - Devinder S Arora
- School of Pharmacy, Griffith University Southport, QLD, Australia
| | - Maureen A McElvain
- Department of Physiology and Biophysics, University of Illinois at Chicago Chicago, IL, USA
| | - Bertha J Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago Chicago, IL, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago Chicago, IL, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
44
|
Huynh N, Arabian N, Naito A, Louie S, Jakowec MW, Asatryan L, Davies DL. Preclinical development of moxidectin as a novel therapeutic for alcohol use disorder. Neuropharmacology 2016; 113:60-70. [PMID: 27641072 DOI: 10.1016/j.neuropharm.2016.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/14/2016] [Accepted: 09/15/2016] [Indexed: 02/08/2023]
Abstract
Current pharmacotherapies for alcohol used disorder (AUD) are few and relatively ineffective illustrating the need for the development of new, effective medications. Using a translational approach, our laboratory reported that ivermectin, an FDA-approved, human and animal anti-parasitic agent, can significantly reduce ethanol intake in male and female mice across different drinking paradigms. Extending this line of investigation, the current paper investigated the utility of moxidectin (MOX), an analogue of ivermectin, to reduce ethanol intake. Notably, MOX is widely held to have lower neurotoxicity potential and improved margin of safety compared to ivermectin. Using a 24-h-two-bottle choice paradigm, MOX significantly reduced ethanol intake in a dose dependent manner in both male and female C57BL/6J mice, respectively (1.25-7.5 mg/kg) and (1.25-10 mg/kg). Further, multi-day administration of MOX (2.5 mg/kg; intraperitoneal injection) for 5 consecutive days significantly reduced ethanol intake in both the 24-h-two-bottle choice and Drinking-in-the-Dark paradigms in female mice. No overt signs of behavioral toxicity were observed. Notably in both male and female mice, MOX significantly reduced ethanol intake starting approximately 4 h post-injection. Using a Xenopus oocyte expression system, we found that MOX significantly potentiated P2X4 receptor (P2X4R) function and antagonized the inhibitory effects of ethanol on ATP-gated currents in P2X4Rs. This latter finding represents the first report of MOX having activity on P2X4Rs. In addition, MOX potentiated GABAA receptors, but to a lesser degree as compared to ivermectin supporting the hypothesis that MOX would be advantageous (compared to ivermectin) with respect to reducing contraindications. Overall, the results illustrate the potential for development of MOX as a novel pharmacotherapy for the treatment of AUD.
Collapse
Affiliation(s)
- Nhat Huynh
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Natalie Arabian
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Anna Naito
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Stan Louie
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA.
| |
Collapse
|
45
|
Ahlers KE, Chakravarti B, Fisher RA. RGS6 as a Novel Therapeutic Target in CNS Diseases and Cancer. AAPS JOURNAL 2016; 18:560-72. [PMID: 27002730 DOI: 10.1208/s12248-016-9899-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/25/2016] [Indexed: 12/17/2022]
Abstract
Regulator of G protein signaling (RGS) proteins are gatekeepers regulating the cellular responses induced by G protein-coupled receptor (GPCR)-mediated activation of heterotrimeric G proteins. Specifically, RGS proteins determine the magnitude and duration of GPCR signaling by acting as a GTPase-activating protein for Gα subunits, an activity facilitated by their semiconserved RGS domain. The R7 subfamily of RGS proteins is distinguished by two unique domains, DEP/DHEX and GGL, which mediate membrane targeting and stability of these proteins. RGS6, a member of the R7 subfamily, has been shown to specifically modulate Gαi/o protein activity which is critically important in the central nervous system (CNS) for neuronal responses to a wide array of neurotransmitters. As such, RGS6 has been implicated in several CNS pathologies associated with altered neurotransmission, including the following: alcoholism, anxiety/depression, and Parkinson's disease. In addition, unlike other members of the R7 subfamily, RGS6 has been shown to regulate G protein-independent signaling mechanisms which appear to promote both apoptotic and growth-suppressive pathways that are important in its tumor suppressor function in breast and possibly other tissues. Further highlighting the importance of RGS6 as a target in cancer, RGS6 mediates the chemotherapeutic actions of doxorubicin and blocks reticular activating system (Ras)-induced cellular transformation by promoting degradation of DNA (cytosine-5)-methyltransferase 1 (DNMT1) to prevent its silencing of pro-apoptotic and tumor suppressor genes. Together, these findings demonstrate the critical role of RGS6 in regulating both G protein-dependent CNS pathology and G protein-independent cancer pathology implicating RGS6 as a novel therapeutic target.
Collapse
Affiliation(s)
- Katelin E Ahlers
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA
| | - Bandana Chakravarti
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA
| | - Rory A Fisher
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA. .,Department of Internal Medicine, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
46
|
Fritz BM, Boehm SL. Rodent models and mechanisms of voluntary binge-like ethanol consumption: Examples, opportunities, and strategies for preclinical research. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:297-308. [PMID: 26021391 PMCID: PMC4668238 DOI: 10.1016/j.pnpbp.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 05/02/2015] [Accepted: 05/21/2015] [Indexed: 02/03/2023]
Abstract
Binge ethanol consumption has widespread negative consequences for global public health. Rodent models offer exceptional power to explore the neurobiology underlying and affected by binge-like drinking as well as target potential prevention, intervention, and treatment strategies. An important characteristic of these models is their ability to consistently produce pharmacologically-relevant blood ethanol concentration. This review examines the current available rodent models of voluntary, pre-dependent binge-like ethanol consumption and their utility in various research strategies. Studies have demonstrated that a diverse array of neurotransmitters regulate binge-like drinking, resembling some findings from other drinking models. Furthermore, repeated binge-like drinking recruits neuroadaptive mechanisms in mesolimbocortical reward circuitry. New opportunities that these models offer in the current context of mechanistic research are also discussed.
Collapse
Affiliation(s)
| | - Stephen L Boehm
- Indiana Alcohol Research Center, Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States.
| |
Collapse
|
47
|
West CHK, Boss-Williams KA, Ritchie JC, Weiss JM. Reprint of: Locus coeruleus neuronal activity determines proclivity to consume alcohol in a selectively-bred line of rats that readily consumes alcohol. Alcohol 2016; 50:91-105. [PMID: 26873226 DOI: 10.1016/j.alcohol.2016.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/21/2015] [Accepted: 08/18/2015] [Indexed: 11/18/2022]
Abstract
Sprague-Dawley rats selectively-bred for susceptibility to stress in our laboratory (Susceptible, or SUS rats) voluntarily consume large amounts of alcohol, and amounts that have, as shown here, pharmacological effects, which normal rats will not do. In this paper, we explore neural events in the brain that underlie this propensity to readily consume alcohol. Activity of locus coeruleus neurons (LC), the major noradrenergic cell body concentration in the brain, influences firing of ventral tegmentum dopaminergic cell bodies of the mesocorticolimbic system (VTA-DA neurons), which mediate rewarding aspects of alcohol. We tested the hypothesis that in SUS rats alcohol potently suppresses LC activity to markedly diminish LC-mediated inhibition of VTA-DA neurons, which permits alcohol to greatly increase VTA-DA activity and rewarding aspects of alcohol. Electrophysiological single-unit recording of LC and VTA-DA activity showed that in SUS rats alcohol decreased LC burst firing much more than in normal rats and as a result markedly increased VTA-DA activity in SUS rats while having no such effect in normal rats. Consistent with this, in a behavioral test for reward using conditioned place preference (CPP), SUS rats showed alcohol, given by intraperitoneal (i.p.) injection, to be rewarding. Next, manipulation of LC activity by microinfusion of drugs into the LC region of SUS rats showed that (a) decreasing LC activity increased alcohol intake and increasing LC activity decreased alcohol intake in accord with the formulation described above, and (b) increasing LC activity blocked both the rewarding effect of alcohol in the CPP test and the usual alcohol-induced increase in VTA-DA single-unit activity seen in SUS rats. An important ancillary finding in the CPP test was that an increase in LC activity was rewarding by itself, while a decrease in LC activity was aversive; consequently, effects of LC manipulations on alcohol-related reward in the CPP test were perhaps even larger than evident in the test. Finally, when increased LC activity was associated with (i.e., conditioned to) i.p. alcohol, subsequent alcohol consumption by SUS rats was markedly reduced, indicating that SUS rats consume large amounts of alcohol because of rewarding physiological consequences requiring increased VTA-DA activity. The findings reported here are consistent with the view that the influence of alcohol on LC activity leading to changes in VTA-DA activity strongly affects alcohol-mediated reward, and may well be the basis of the proclivity of SUS rats to avidly consume alcohol.
Collapse
Affiliation(s)
- Charles H K West
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Katherine A Boss-Williams
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - James C Ritchie
- Department of Pathology, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Jay M Weiss
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|
48
|
Bell RL, Hauser SR, McClintick J, Rahman S, Edenberg HJ, Szumlinski KK, McBride WJ. Ethanol-Associated Changes in Glutamate Reward Neurocircuitry: A Minireview of Clinical and Preclinical Genetic Findings. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:41-85. [PMID: 26809998 DOI: 10.1016/bs.pmbts.2015.10.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Herein, we have reviewed the role of glutamate, the major excitatory neurotransmitter in the brain, in a number of neurochemical, -physiological, and -behavioral processes mediating the development of alcohol dependence. The findings discussed include results from both preclinical as well as neuroimaging and postmortem clinical studies. Expression levels for a number of glutamate-associated genes and/or proteins are modulated by alcohol abuse and dependence. These changes in expression include metabotropic receptors and ionotropic receptor subunits as well as different glutamate transporters. Moreover, these changes in gene expression parallel the pharmacologic manipulation of these same receptors and transporters. Some of these gene expression changes may have predated alcohol abuse and dependence because a number of glutamate-associated polymorphisms are related to a genetic predisposition to develop alcohol dependence. Other glutamate-associated polymorphisms are linked to age at the onset of alcohol-dependence and initial level of response/sensitivity to alcohol. Finally, findings of innate and/or ethanol-induced glutamate-associated gene expression differences/changes observed in a genetic animal model of alcoholism, the P rat, are summarized. Overall, the existing literature indicates that changes in glutamate receptors, transporters, enzymes, and scaffolding proteins are crucial for the development of alcohol dependence and there is a substantial genetic component to these effects. This indicates that continued research into the genetic underpinnings of these glutamate-associated effects will provide important novel molecular targets for treating alcohol abuse and dependence.
Collapse
Affiliation(s)
- Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | - Sheketha R Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jeanette McClintick
- Departments of Biochemistry and Molecular Biology and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana , USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Howard J Edenberg
- Departments of Biochemistry and Molecular Biology and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana , USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California, USA
| | - William J McBride
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
49
|
Silberman Y, Fetterly TL, Awad EK, Milano EJ, Usdin TB, Winder DG. Ethanol produces corticotropin-releasing factor receptor-dependent enhancement of spontaneous glutamatergic transmission in the mouse central amygdala. Alcohol Clin Exp Res 2015; 39:2154-62. [PMID: 26503065 PMCID: PMC4624256 DOI: 10.1111/acer.12881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/14/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ethanol (EtOH) modulation of central amygdala (CeA) neurocircuitry plays a key role in the development of alcoholism via activation of the corticotropin-releasing factor (CRF) receptor (CRFR) system. Previous work has predominantly focused on EtOH × CRF interactions on the CeA GABA circuitry; however, our laboratory recently showed that CRF enhances CeA glutamatergic transmission. Therefore, this study sought to determine whether EtOH modulates CeA glutamate transmission via activation of CRF signaling. METHODS The effects of EtOH on spontaneous excitatory postsynaptic currents (sEPSCs) and basal resting membrane potentials were examined via standard electrophysiology methods in adult male C57BL/6J mice. Local ablation of CeA CRF neurons (CRF(CeAhDTR) ) was achieved by targeting the human diphtheria toxin receptor (hDTR) to CeA CRF neurons with an adeno-associated virus. Ablation was quantified post hoc with confocal microscopy. Genetic targeting of the diphtheria toxin active subunit to CRF neurons (CRF(DTA) mice) ablated CRF neurons throughout the central nervous system, as assessed by quantitative reverse transcriptase polymerase chain reaction quantification of CRF mRNA. RESULTS Acute bath application of EtOH significantly increased sEPSC frequency in a concentration-dependent manner in CeA neurons, and this effect was blocked by pretreatment of co-applied CRFR1 and CRFR2 antagonists. In experiments utilizing a CRF-tomato reporter mouse, EtOH did not significantly alter the basal membrane potential of CeA CRF neurons. The ability of EtOH to enhance CeA sEPSC frequency was not altered in CRF(CeAhDTR) mice despite a ~78% reduction in CeA CRF cell counts. The ability of EtOH to enhance CeA sEPSC frequency was also not altered in the CRF(DTA) mice despite a 3-fold reduction in CRF mRNA levels. CONCLUSIONS These findings demonstrate that EtOH enhances spontaneous glutamatergic transmission in the CeA via a CRFR-dependent mechanism. Surprisingly, our data suggest that this action may not require endogenous CRF.
Collapse
Affiliation(s)
- Yuval Silberman
- Molecular Physiology and Biophysics, Neuroscience Program, Vanderbilt University School of Medicine
| | - Tracy L. Fetterly
- Molecular Physiology and Biophysics, Neuroscience Program, Vanderbilt University School of Medicine
| | - Elias K. Awad
- Molecular Physiology and Biophysics, Neuroscience Program, Vanderbilt University School of Medicine
| | - Elana J. Milano
- Molecular Physiology and Biophysics, Neuroscience Program, Vanderbilt University School of Medicine
| | - Ted B. Usdin
- Section Fundamental Neuroscience, National Institute of Mental Health, National Institutes of Health
| | - Danny G. Winder
- Molecular Physiology and Biophysics, Neuroscience Program, Vanderbilt University School of Medicine
| |
Collapse
|
50
|
West CHK, Boss-Williams KA, Ritchie JC, Weiss JM. Locus coeruleus neuronal activity determines proclivity to consume alcohol in a selectively-bred line of rats that readily consumes alcohol. Alcohol 2015; 49:691-705. [PMID: 26496795 DOI: 10.1016/j.alcohol.2015.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/21/2015] [Accepted: 08/18/2015] [Indexed: 02/02/2023]
Abstract
Sprague-Dawley rats selectively-bred for susceptibility to stress in our laboratory (Susceptible, or SUS rats) voluntarily consume large amounts of alcohol, and amounts that have, as shown here, pharmacological effects, which normal rats will not do. In this paper, we explore neural events in the brain that underlie this propensity to readily consume alcohol. Activity of locus coeruleus neurons (LC), the major noradrenergic cell body concentration in the brain, influences firing of ventral tegmentum dopaminergic cell bodies of the mesocorticolimbic system (VTA-DA neurons), which mediate rewarding aspects of alcohol. We tested the hypothesis that in SUS rats alcohol potently suppresses LC activity to markedly diminish LC-mediated inhibition of VTA-DA neurons, which permits alcohol to greatly increase VTA-DA activity and rewarding aspects of alcohol. Electrophysiological single-unit recording of LC and VTA-DA activity showed that in SUS rats alcohol decreased LC burst firing much more than in normal rats and as a result markedly increased VTA-DA activity in SUS rats while having no such effect in normal rats. Consistent with this, in a behavioral test for reward using conditioned place preference (CPP), SUS rats showed alcohol, given by intraperitoneal (i.p.) injection, to be rewarding. Next, manipulation of LC activity by microinfusion of drugs into the LC region of SUS rats showed that (a) decreasing LC activity increased alcohol intake and increasing LC activity decreased alcohol intake in accord with the formulation described above, and (b) increasing LC activity blocked both the rewarding effect of alcohol in the CPP test and the usual alcohol-induced increase in VTA-DA single-unit activity seen in SUS rats. An important ancillary finding in the CPP test was that an increase in LC activity was rewarding by itself, while a decrease in LC activity was aversive; consequently, effects of LC manipulations on alcohol-related reward in the CPP test were perhaps even larger than evident in the test. Finally, when increased LC activity was associated with (i.e., conditioned to) i.p. alcohol, subsequent alcohol consumption by SUS rats was markedly reduced, indicating that SUS rats consume large amounts of alcohol because of rewarding physiological consequences requiring increased VTA-DA activity. The findings reported here are consistent with the view that the influence of alcohol on LC activity leading to changes in VTA-DA activity strongly affects alcohol-mediated reward, and may well be the basis of the proclivity of SUS rats to avidly consume alcohol.
Collapse
Affiliation(s)
- Charles H K West
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Katherine A Boss-Williams
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - James C Ritchie
- Department of Pathology, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Jay M Weiss
- Department of Psychiatry and Behavioral Sciences, Emory University, School of Medicine, Woodruff Memorial Research Building (WMB), 4th Floor, 101 Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|