1
|
Barrios N, Riordan W, Garcia-Rivas V, Peltier MR, Roberts W, Verplaetse TL, Kohler R, Zhou H, Banini BA, McKee SA, Cosgrove KP, Zakiniaeiz Y. Preclinical and clinical sex differences in the effects of alcohol on measures of brain dopamine: a systematic review. Biol Sex Differ 2025; 16:24. [PMID: 40200334 PMCID: PMC11980350 DOI: 10.1186/s13293-025-00706-7] [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: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
INTRODUCTION Dopamine is involved in reward processing and plays a critical role in the development and progression of alcohol use disorder (AUD). However, little is known about the effect of sex on the relationship between dopamine and alcohol use/AUD. There is a critical need to identify the neurobiological mechanisms that contribute to sex differences in AUD to inform treatment approaches. This study aimed to review existing literature on sex differences in the effects of alcohol on brain dopamine measures in animals and individuals with heavy drinking/AUD. METHODS A systematic review was conducted using Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. PubMed was searched from inception to July 23rd, 2024. RESULTS Of the 1,412 articles identified, 10 met study criteria (1 human, 9 animal), including in vivo (two positron emission tomography, four microdialysis) and ex vivo (two liquid chromatography, two fast-scan cyclic voltammetry) studies. Six studies included an alcohol challenge; three showed that females had greater alcohol-induced dopamine release than males in the ventral striatum and frontal cortex, while three showed no sex-related differences. Notably, the latter three studies examined sex in a combined AUD/control group or measured dopamine levels days after alcohol exposure. Two studies that examined the effects of prenatal alcohol exposure showed that prenatal-alcohol-exposed male offspring versus sex-matched air-exposed controls had greater prefrontal cortical dopamine D1 receptor availability, and prenatal-alcohol-exposed female offspring versus sex-matched air-exposed controls had greater striatal dopamine concentration. Two studies investigating the mu-opioid receptor (MOR) regulation of alcohol-induced dopamine release showed a faster decline in females relative to males while the other study found females may be less dependent on MOR activity at lower doses of alcohol relative to higher doses. CONCLUSIONS This systematic review showed mixed results regarding sex differences in brain dopamine measures in alcohol-exposed animals and individuals with AUD, which may arise from differences in the timing, quantity, and duration of alcohol exposure, species, conditions, models, and techniques. More research examining the effect of sex on the relationship between alcohol use and brain dopamine measures is needed to enhance our understanding of AUD development, progression, and treatment in both females and males.
Collapse
Affiliation(s)
- Nathalie Barrios
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Will Riordan
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - MacKenzie R Peltier
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- Psychology Service, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Walter Roberts
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Terril L Verplaetse
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Robert Kohler
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Hang Zhou
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Bubu A Banini
- Section of Digestive Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, USA
| | - Sherry A McKee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Kelly P Cosgrove
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- Yale Positron Emission Tomography (PET) Center, School of Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, USA
| | - Yasmin Zakiniaeiz
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA.
- , 40 Temple Street, Suite 7C, New Haven, CT, 06519, USA.
| |
Collapse
|
2
|
Harris BB, Sinha R, Goldfarb EV. Drawbacks to Strengthening Neural Salience Encoding: A Link Between Cortisol and Risky Drinking. J Neurosci 2024; 44:e1027242024. [PMID: 39147591 PMCID: PMC11450530 DOI: 10.1523/jneurosci.1027-24.2024] [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: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Emotionally salient experiences are encoded and remembered more strongly, an effect that can be amplified by hormones like cortisol. Such memories can in turn profoundly influence later behavior. However, little is known about the link between amplified salience encoding and subsequent behavior. This pathway may be particularly important for risky alcohol drinking, which has been linked to sensitized salience responses, memory, and cortisol. To test this possibility, we integrated pharmacology using a double-blind cross-over design with fMRI, cognitive, and motivation assays across a range of healthy male and female social drinkers. As anticipated, cortisol enhanced memory for salient alcohol-related events; critically, this bias was in turn associated with later alcohol motivation. Increased alcohol motivation was particularly pronounced in more susceptible risky drinkers, for whom cortisol enhanced brain salience responses to alcohol. These sensitized salience responses predicted both memory biases and alcohol motivation. Together, these findings reveal maladaptive consequences of enhanced salience encoding.
Collapse
Affiliation(s)
- Bailey B Harris
- Department of Psychology, UCLA, Los Angeles, California 90095
| | - Rajita Sinha
- Department of Psychiatry, Yale University, New Haven, Connecticut 06511
| | - Elizabeth V Goldfarb
- Department of Psychiatry, Yale University, New Haven, Connecticut 06511
- Department of Psychology, Yale University, New Haven, Connecticut 06520
- Wu Tsai Institute, Yale University, New Haven, Connecticut 06510
- National Center for PTSD, West Haven, Connecticut 06477
| |
Collapse
|
3
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [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: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
4
|
Besong OTO, Koo JS, Zhang H. Brain lncRNA-mRNA co-expression regulatory networks and alcohol use disorder. Genomics 2024; 116:110928. [PMID: 39197730 PMCID: PMC11531342 DOI: 10.1016/j.ygeno.2024.110928] [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: 03/12/2024] [Revised: 08/08/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
Prolonged alcohol consumption can disturb the expression of both coding and noncoding genes in the brain. These dysregulated genes may co-express in modules and interact within networks, consequently influencing the susceptibility to developing alcohol use disorder (AUD). In the present study, we performed an RNA-seq analysis of the expression of both long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) in 192 postmortem tissue samples collected from eight brain regions (amygdala, caudate nucleus, cerebellum, hippocampus, nucleus accumbens, prefrontal cortex, putamen, and ventral tegmental area) of 12 AUD and 12 control subjects of European ancestry. Applying the limma-voom method, we detected a total of 57 lncRNAs and 51 mRNAs exhibiting significant differential expression (Padj < 0.05 and fold-change ≥2) across at least one of the eight brain regions investigated. Machine learning analysis further confirmed the potential of these top genes in predicting AUD. Through Weighted Gene Co-expression Network Analysis (WGCNA), we identified distinct lncRNA-mRNA co-expression modules associated with AUD in each of the eight brain regions. Additionally, lncRNA-mRNA co-expression networks were constructed for each brain region using Cytoscape to reveal gene regulatory interactions implicated in AUD. Hub genes within these networks were found to be enriched in several key KEGG pathways, including Axon Guidance, MAPK Signaling, p53 Signaling, Adherens Junction, and Neurodegeneration. Our results underscore the significance of networks involving AUD-associated lncRNAs and mRNAs in modulating neuroplasticity in response to alcohol exposure. Further elucidating these molecular mechanisms holds promise for the development of targeted therapeutic interventions for AUD.
Collapse
Affiliation(s)
- Ojong Tabi Ojong Besong
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ji Sun Koo
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
5
|
Vierkant V, Xie X, Huang Z, He L, Bancroft E, Wang X, Nguyen T, Srinivasan R, Zhou Y, Wang J. Optogenetic inhibition of light-captured alcohol-taking striatal engrams facilitates extinction and suppresses reinstatement. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1728-1739. [PMID: 39095328 PMCID: PMC11576255 DOI: 10.1111/acer.15412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a complex condition, and it remains unclear which specific neuronal substrates mediate alcohol-seeking and -taking behaviors. Engram cells and their related ensembles, which encode learning and memory, may play a role in this process. We aimed to assess the precise neural substrates underlying alcohol-seeking and -taking behaviors and determine how they may affect one another. METHODS Using FLiCRE (Fast Light and Calcium-Regulated Expression; a newly developed technique which permits the trapping of acutely activated neuronal ensembles) and operant self-administration (OSA), we tagged striatal neurons activated during alcohol-taking behaviors. We used FLiCRE to express an inhibitory halorhodopsin in alcohol-taking neurons, permitting loss-of-function manipulations. RESULTS We found that the inhibition of OSA-tagged alcohol-taking neurons decreased both alcohol-seeking and -taking behaviors in future OSA trials. In addition, optogenetic inhibition of these OSA-tagged alcohol-taking neurons during extinction training facilitated the extinction of alcohol-seeking behaviors. Furthermore, inhibition of these OSA-tagged alcohol-taking neurons suppressed the reinstatement of alcohol-seeking behaviors, but, interestingly, it did not significantly suppress alcohol-taking behaviors during reinstatement. CONCLUSIONS Our findings suggest that alcohol-taking neurons are crucial for future alcohol-seeking behaviors during extinction and reinstatement. These results may help in the development of new therapeutic approaches to enhance extinction and suppress relapse in individuals with AUD.
Collapse
Affiliation(s)
- Valerie Vierkant
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Zhenbo Huang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Eric Bancroft
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Tran Nguyen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
6
|
Leclercq S. Involvement of the gut microbiome-brain axis in alcohol use disorder. Alcohol Alcohol 2024; 59:agae050. [PMID: 39042929 DOI: 10.1093/alcalc/agae050] [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: 04/18/2024] [Revised: 06/06/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
The human intestine is colonized by a variety of microorganisms that influence the immune system, the metabolic response, and the nervous system, with consequences for brain function and behavior. Unbalance in this microbial ecosystem has been shown to be associated with psychiatric disorders, and altered gut microbiome composition related to bacteria, viruses, and fungi has been well established in patients with alcohol use disorder. This review describes the gut microbiome-brain communication pathways, including the ones related to the vagus nerve, the inflammatory cytokines, and the gut-derived metabolites. Finally, the potential benefits of microbiota-based therapies for the management of alcohol use disorder, such as probiotics, prebiotics, and fecal microbiota transplantation, are also discussed.
Collapse
Affiliation(s)
- Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
7
|
Cristina Bianchi P, Palombo P, Antonagi Engi S, Eduardo Carneiro de Oliveira P, Emily Boaventura Tavares G, Anjos-Santos A, Suemi Yokoyama T, da Silva Planeta C, Cardoso Cruz F, Molini Leão R. Involvement of Pre-limbic Cortex-Nucleus accumbens projections in Context-Induced alcohol seeking. Brain Res 2024; 1841:149086. [PMID: 38876319 DOI: 10.1016/j.brainres.2024.149086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Alcohol use disorder (AUD) remains a critical public health issue worldwide, characterized by high relapse rates often triggered by contextual cues. This research investigates the neural mechanisms behind context-induced reinstatement of alcohol-seeking behavior, focusing on the nucleus accumbens and its interactions with the prelimbic cortex, employing Male Long-Evans rats in an ABA renewal model. In our experimental setup, rats were trained to self-administer 10 % ethanol in Context A, followed by extinction of lever pressing in the presence of discrete cues in Context B. The context-induced reinstatement of ethanol-seeking was then assessed by re-exposing rats to Context A or B under extinction conditions, aiming to simulate the environmental cues' influence on relapse behaviors. Three experiments were conducted: Experiment 1 utilized Fos-immunohistochemistry to examine neuronal activation in the nucleus accumbens; Experiment 2 applied the baclofen + muscimol inactivation technique to probe the functional importance of the nucleus accumbens core; Experiment 3 used Fos-immunofluorescence along with Retrobeads injection to investigate activation of neurons projecting from the prelimbic cortex to the nucleus accumbens core. Our findings revealed significant increases in Fos-immunoreactive nuclei within the nucleus accumbens core and shell during the reinstatement phase in Context A, underscoring the environment's potent effect on ethanol-seeking behavior. Additionally, inactivation of the nucleus accumbens core markedly reduced reinstatement, and there was a notable activation of neurons from the prelimbic cortex to the nucleus accumbens core in the ethanol-associated context. These results highlight the critical role of the nucleus accumbens core and its corticostriatal projections in the neural circuitry underlying context-driven ethanol seeking.
Collapse
Affiliation(s)
- Paula Cristina Bianchi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Paola Palombo
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Sheila Antonagi Engi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | | | | | - Alexia Anjos-Santos
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Thais Suemi Yokoyama
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Cleopatra da Silva Planeta
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, UNESP, Araraquara, SP, Brazil
| | - Fabio Cardoso Cruz
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Rodrigo Molini Leão
- Laboratory of Pharmacology, Biomedical Sciences Institute, Department of Pharmacology, Federal University of Uberlândia, Uberlândia, MG, Brazil; Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia/MG, Brazil.
| |
Collapse
|
8
|
Vierkant V, Xie X, Huang Z, He L, Bancroft E, Wang X, Srinivisan R, Zhou Y, Wang J. Optogenetic inhibition of light-captured alcohol-taking striatal engrams facilitates extinction and suppresses reinstatement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597021. [PMID: 38853893 PMCID: PMC11160798 DOI: 10.1101/2024.06.02.597021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Alcohol use disorder (AUD) is a complex condition, and it remains unclear which specific neuronal substrates mediate alcohol-seeking and -taking behaviors. Engram cells and their related ensembles, which encode learning and memory, may play a role in this process. We aimed to assess the precise neural substrates underlying alcohol-seeking and -taking behaviors and determine how they may affect one another. Methods Using FLiCRE (Fast Light and Calcium-Regulated Expression; a newly developed technique which permits the trapping of acutely activated neuronal ensembles) and operant-self administration (OSA), we tagged striatal neurons activated during alcohol-taking behaviors. We used FLiCRE to express an inhibitory halorhodopsin in alcohol-taking neurons, permitting loss-of-function manipulations. Results We found that the inhibition of OSA-tagged alcohol-taking neurons decreased both alcohol-seeking and -taking behaviors in future OSA trials. In addition, optogenetic inhibition of these OSA-tagged alcohol-taking neurons during extinction training facilitated the extinction of alcohol-seeking behaviors. Furthermore, inhibition of these OSA-tagged alcohol-taking neurons suppressed the reinstatement of alcohol-seeking behaviors, but, interestingly, it did not significantly suppress alcohol-taking behaviors during reinstatement. Conclusions Our findings suggest that alcohol-taking neurons are crucial for future alcohol-seeking behaviors during extinction and reinstatement. These results may help in the development of new therapeutic approaches to enhance extinction and suppress relapse in individuals with AUD.
Collapse
|
9
|
Kale MB, Chandurkar PA, Taksande BG, Aglawe MM, Rahangdale SR, Upaganlawar AB, Kopalli SR, Umekar MJ, Wankhede NL. Agmatine alleviates ethanol withdrawal-associated cognitive impairment and neurochemical imbalance in rats. Neurosci Lett 2024; 832:137804. [PMID: 38692559 DOI: 10.1016/j.neulet.2024.137804] [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: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
The present study aimed to investigate the role of agmatine in the neurobiology underlying memory impairment during ethanol withdrawal in rats. Sprague-Dawley rats were subjected to a 21-day chronic ethanol exposure regimen (2.4 % w/v ethanol for 3 days, 4.8 % w/v for the next 4 days, and 7.2 % w/v for the following 14 days), followed by a withdrawal period. Memory impairment was assessed using the passive avoidance test (PAT) at 24, 48, and 72 h post-withdrawal. The ethanol-withdrawn rats displayed a significant decrease in step-through latency in the PAT, indicative of memory impairment at 72 h post-withdrawal. However, administration of agmatine (40 µg/rat) and its modulators (L-arginine, arcaine, and amino-guanidine) significantly increases the latency time in the ethanol-withdrawn rats, demonstrating the attenuation of memory impairment. Further, pretreatment with imidazoline receptor agonists enhances agmatine's effects, while antagonists block them, implicating imidazoline receptors in agmatine's actions. Neurochemical analysis in ethanol-withdrawn rats reveals dysregulated glutamate and GABA levels, which was attenuated by agmatine and its modulators. By examining the effects of agmatine administration and modulators of endogenous agmatine, the study aimed to shed light on the potential therapeutic implications of agmatinergic signaling in alcohol addiction and related cognitive deficits. Thus, the present findings suggest that agmatine administration and modulation of endogenous agmatine levels hold potential as therapeutic strategies for managing alcohol addiction and associated cognitive deficits. Understanding the neurobiology underlying these effects paves the way for the development of novel interventions targeting agmatinergic signaling in addiction treatment.
Collapse
Affiliation(s)
- Mayur B Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India; SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| | - Pranali A Chandurkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh G Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Manish M Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Sandip R Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| | - Spandana R Kopalli
- Department of Integrated Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Milind J Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Nitu L Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India; SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| |
Collapse
|
10
|
Dilly GA, Blednov YA, Warden AS, Ezerskiy L, Fleischer C, Plotkin JD, Patil S, Osterndorff-Kahanek EA, Mayfield J, Mayfield RD, Homanics GE, Messing RO. Knockdown of Tlr3 in dorsal striatum reduces ethanol consumption and acute functional tolerance in male mice. Brain Behav Immun 2024; 118:437-448. [PMID: 38499210 PMCID: PMC11007683 DOI: 10.1016/j.bbi.2024.03.021] [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: 08/16/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Systemic activation of toll-like receptor 3 (TLR3) signaling using poly(I:C), a TLR3 agonist, drives ethanol consumption in several rodent models, while global knockout of Tlr3 reduces drinking in C57BL/6J male mice. To determine if brain TLR3 pathways are involved in drinking behavior, we used CRISPR/Cas9 genome editing to generate a Tlr3 floxed (Tlr3F/F) mouse line. After sequence confirmation and functional validation of Tlr3 brain transcripts, we injected Tlr3F/F male mice with an adeno-associated virus expressing Cre recombinase (AAV5-CMV-Cre-GFP) to knockdown Tlr3 in the medial prefrontal cortex, nucleus accumbens, or dorsal striatum (DS). Only Tlr3 knockdown in the DS decreased two-bottle choice, every-other-day (2BC-EOD) ethanol consumption. DS-specific deletion of Tlr3 also increased intoxication and prevented acute functional tolerance to ethanol. In contrast, poly(I:C)-induced activation of TLR3 signaling decreased intoxication in male C57BL/6J mice, consistent with its ability to increase 2BC-EOD ethanol consumption in these mice. We also found that TLR3 was highly colocalized with DS neurons. AAV5-Cre transfection occurred predominantly in neurons, but there was minimal transfection in astrocytes and microglia. Collectively, our previous and current studies show that activating or inhibiting TLR3 signaling produces opposite effects on acute responses to ethanol and on ethanol consumption. While previous studies, however, used global knockout or systemic TLR3 activation (which alter peripheral and brain innate immune responses), the current results provide new evidence that brain TLR3 signaling regulates ethanol drinking. We propose that activation of TLR3 signaling in DS neurons increases ethanol consumption and that a striatal TLR3 pathway is a potential target to reduce excessive drinking.
Collapse
Affiliation(s)
- Geoffrey A Dilly
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Lubov Ezerskiy
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Caleb Fleischer
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jesse D Plotkin
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | | | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Gregg E Homanics
- Departments of Anesthesiology & Perioperative Medicine, Neurobiology, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
11
|
Reguilón MD, Manzanedo C, Miñarro J, Rodríguez-Arias M. Stress inoculation during adolescence attenuates social stress-induced increase in ethanol intake in adult male mice. Neuropharmacology 2024; 246:109838. [PMID: 38199295 DOI: 10.1016/j.neuropharm.2024.109838] [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: 10/10/2023] [Revised: 12/05/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Social stress exposure heightens the risk of substance abuse disorder development, especially when endured during adolescence, influencing long-term mental health. This study investigates early-life stress's potential to confer resilience against later-life stressors. To investigate this hypothesis, we examined the impact of a single social defeat (SD) incident during adolescent mice's lives on subsequent voluntary ethanol consumption following repeated adult social stress exposure. Half of the adolescent mice experienced SD at postnatal day 28. Three weeks later (postnatal day 49), defeated groups encountered four confrontations with aggressive residents every 72 h, while control groups were exposed to non-resident exploration. A day after the last SD, defeated mice were classified as resilient or susceptible based on their response to a social interaction test (SIT), a model for depressive behavior. To assess ethanol consumption during young adulthood, researchers used the 'drinking in the dark' and oral ethanol self-administration paradigms. Stress inoculation (IS) slightly increased resilient animals in the SIT. In mice without IS exposure during adolescence, susceptible defeated mice displayed higher ethanol consumption and motivation than control and resilient mice. IS in adolescence effectively counteracted this effect, as IS-SD groups, whether resilient or susceptible, showed no increase in ethanol intake. These groups also exhibited similar motivation to control, measured by the progressive ratio. Notably, elevated IL-6 levels seen in SD-S mice were absent in IS-exposed mice. Additionally, IS-exposed groups had lower prefrontal cortex IL-6 and CX3CL1 levels. These findings support the hypothesis that IS, induced by moderate-intensity stress during adolescence, can enhance resilience to more severe stressors in adulthood.
Collapse
Affiliation(s)
- Marina D Reguilón
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Carmen Manzanedo
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain.
| |
Collapse
|
12
|
Patton MS, Sheats SH, Siclair AN, Mathur BN. Alcohol potentiates multiple GABAergic inputs to dorsal striatum fast-spiking interneurons. Neuropharmacology 2023; 232:109527. [PMID: 37011784 PMCID: PMC10122715 DOI: 10.1016/j.neuropharm.2023.109527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023]
Abstract
Parvalbumin-expressing dorsal striatal fast-spiking interneurons, comprising ∼1% of the total dorsal striatal neuronal population, are necessary for the expression of compulsive-like ethanol consumption mice. Fast-spiking interneurons are driven to fire by glutamatergic inputs derived primarily from the cortex. However, these neurons also receive substantial GABAergic input from two sources: the globus pallidus and the reticular nucleus of the thalamus. How ethanol modulates inhibitory input onto fast-spiking neurons is unclear and, more broadly, alcohol effects on GABAergic synaptic transmission onto GABAergic interneurons are understudied. Examining this, we found that acute bath application of ethanol (50 mM) potentiated GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto fast-spiking interneurons in mouse of both sexes. This ethanol-induced potentiation required postsynaptic calcium and was not accompanied by a sustained change in presynaptic GABA release probability. Examining whether this ethanol effect persisted following chronic intermittent ethanol exposure, we found attenuated acute-ethanol potentiation of GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto striatal fast-spiking interneurons. These data underscore the impact of ethanol on GABAergic signaling in the dorsal striatum and support the notion that ethanol may disinhibit the dorsolateral striatum.
Collapse
Affiliation(s)
- Michael S Patton
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Samuel H Sheats
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Allison N Siclair
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
13
|
Zeng J, You L, Yang F, Luo Y, Yu S, Yan J, Liu M, Yang X. A meta-analysis of the neural substrates of monetary reward anticipation and outcome in alcohol use disorder. Hum Brain Mapp 2023; 44:2841-2861. [PMID: 36852619 PMCID: PMC10089105 DOI: 10.1002/hbm.26249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/23/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
The capacity to anticipate and detect rewarding outcomes is fundamental for the development of adaptive decision-making and goal-oriented behavior. Delineating the neural correlates of different stages of reward processing is imperative for understanding the neurobiological mechanism underlying alcohol use disorder (AUD). To examine the neural correlates of monetary anticipation and outcome in AUD patients, we performed two separate voxel-wise meta-analyses of functional neuroimaging studies, including 12 studies investigating reward anticipation and 7 studies investigating reward outcome using the monetary incentive delay task. During the anticipation stage, AUD patients displayed decreased activation in response to monetary cues in mesocortical-limbic circuits and sensory areas, including the ventral striatum (VS), insula, hippocampus, inferior occipital gyrus, supramarginal gyrus, lingual gyrus and fusiform gyrus. During the outcome stage, AUD patients exhibited reduced activation in the dorsal striatum, VS and insula, and increased activation in the orbital frontal cortex and medial temporal area. Our findings suggest that different activation patterns are associated with nondrug rewards during different reward processing stages, potentially reflecting a changed sensitivity to monetary reward in AUD.
Collapse
Affiliation(s)
- Jianguang Zeng
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Lantao You
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Fan Yang
- Department of Ultrasonography, West China Second University HospitalSichuan UniversityChengduChina
- Chengdu Chenghua District Maternal and Child Health HospitalSichuan UniversityChengduChina
| | - Ya Luo
- Department of Psychiatry, State Key Lab of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Shuxian Yu
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Jiangnan Yan
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Mengqi Liu
- Department of RadiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xun Yang
- School of Public AffairsChongqing UniversityChongqingChina
| |
Collapse
|
14
|
de Guglielmo G, Simpson S, Kimbrough A, Conlisk D, Baker R, Cantor M, Kallupi M, George O. Voluntary and forced exposure to ethanol vapor produces similar escalation of alcohol drinking but differential recruitment of brain regions related to stress, habit, and reward in male rats. Neuropharmacology 2023; 222:109309. [PMID: 36334765 PMCID: PMC10022477 DOI: 10.1016/j.neuropharm.2022.109309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
A major limitation of the most widely used current animal models of alcohol dependence is that they use forced exposure to ethanol including ethanol-containing liquid diet and chronic intermittent ethanol (CIE) vapor to produce clinically relevant blood alcohol levels (BAL) and addiction-like behaviors. We recently developed a novel animal model of voluntary induction of alcohol dependence using ethanol vapor self-administration (EVSA). However, it is unknown whether EVSA leads to an escalation of alcohol drinking per se, and whether such escalation is associated with neuroadaptations in brain regions related to stress, reward, and habit. To address these issues, we compared the levels of alcohol drinking during withdrawal between rats passively exposed to alcohol (CIE) or voluntarily exposed to EVSA and measured the number of Fos+ neurons during acute withdrawal (16 h) in key brain regions important for stress, reward, and habit-related processes. CIE and EVSA rats exhibited similar BAL and similar escalation of alcohol drinking and motivation for alcohol during withdrawal. Acute withdrawal from EVSA and CIE recruited a similar number of Fos+ neurons in the Central Amygdala (CeA), however, acute withdrawal from EVSA recruited a higher number of Fos+ neurons in every other brain region analyzed compared to acute withdrawal from CIE. In summary, while the behavioral measures of alcohol dependence between the voluntary (EVSA) and passive (CIE) model were similar, the recruitment of neuronal ensembles during acute withdrawal was very different. The EVSA model may be particularly useful to unveil the neuronal networks and pharmacology responsible for the voluntary induction and maintenance of alcohol dependence and may improve translational studies by providing preclinical researchers with an animal model that highlights the volitional aspects of alcohol use disorder.
Collapse
Affiliation(s)
| | - Sierra Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47906, USA
| | - Dana Conlisk
- Univ. Bordeaux, INSERM, Neurocenter Magendie, Psychobiology of Drug Addiction Group, U1215, F-33000, Bordeaux, France
| | - Robert Baker
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Maxwell Cantor
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
15
|
Salinas AG, Nadel JA, Mateo Y, Huynh T, Augustin SM, Pacak K, Lovinger DM. Chronic Ethanol Consumption Alters Presynaptic Regulation of Dorsal Striatal Dopamine Release in C57BL/6J Mice. Int J Mol Sci 2022; 23:ijms231910994. [PMID: 36232321 PMCID: PMC9570171 DOI: 10.3390/ijms231910994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized by escalating alcohol consumption, preoccupation with alcohol, and continued alcohol consumption despite adverse consequences. Dopamine has been implicated in neural and behavioral processes involved in reward and reinforcement and is a critical neurotransmitter in AUD. Clinical and preclinical research has shown that long-term ethanol exposure can alter dopamine release, though most of this work has focused on nucleus accumbens (NAc). Like the NAc, the dorsal striatum (DS) is implicated in neural and behavioral processes in AUD. However, little work has examined chronic ethanol effects on DS dopamine dynamics. Therefore, we examined the effect of ethanol consumption and withdrawal on dopamine release and its presynaptic regulation with fast-scan cyclic voltammetry in C57BL/6J mice. We found that one month of ethanol consumption did not alter maximal dopamine release or dopamine tissue content. However, we did find that D2 dopamine autoreceptors were sensitized. We also found a decrease in cholinergic control of dopamine release via β2-containing nAChRs on dopamine axons. Interestingly, both effects were reversed following withdrawal, raising the possibility that some of the neuroadaptations in AUD might be reversible in abstinence. Altogether, this work elucidates some of the chronic alcohol-induced neurobiological dysfunctions in the dopamine system.
Collapse
Affiliation(s)
- Armando G. Salinas
- Laboratory for Integrative Neuroscience, Division of Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Correspondence: (A.G.S.); (D.M.L.)
| | - Jacob A. Nadel
- Laboratory for Integrative Neuroscience, Division of Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA
| | - Yolanda Mateo
- Laboratory for Integrative Neuroscience, Division of Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA
| | - Thanh Huynh
- Section on Medical Neuroendocrinology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shana M. Augustin
- Laboratory for Integrative Neuroscience, Division of Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - David M. Lovinger
- Laboratory for Integrative Neuroscience, Division of Clinical and Biomedical Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20852, USA
- Correspondence: (A.G.S.); (D.M.L.)
| |
Collapse
|
16
|
Zhang J, Song C, Dai J, Li L, Yang X, Chen Z. Mechanism of opioid addiction and its intervention therapy: Focusing on the reward circuitry and mu-opioid receptor. MedComm (Beijing) 2022; 3:e148. [PMID: 35774845 PMCID: PMC9218544 DOI: 10.1002/mco2.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/09/2022] Open
Abstract
Opioid abuse and addiction have become a global pandemic, posing tremendous health and social burdens. The rewarding effects and the occurrence of withdrawal symptoms are the two mainstays of opioid addiction. Mu-opioid receptors (MORs), a member of opioid receptors, play important roles in opioid addiction, mediating both the rewarding effects of opioids and opioid withdrawal syndrome (OWS). The underlying mechanism of MOR-mediated opioid rewarding effects and withdrawal syndrome is of vital importance to understand the nature of opioid addiction and also provides theoretical basis for targeting MORs to treat drug addiction. In this review, we first briefly introduce the basic concepts of MORs, including their structure, distribution in the nervous system, endogenous ligands, and functional characteristics. We focused on the brain circuitry and molecular mechanism of MORs-mediated opioid reward and withdrawal. The neuroanatomical and functional elements of the neural circuitry of the reward system underlying opioid addiction were thoroughly discussed, and the roles of MOR within the reward circuitry were also elaborated. Furthermore, we interrogated the roles of MORs in OWS, along with the structural basis and molecular adaptions of MORs-mediated withdrawal syndrome. Finally, current treatment strategies for opioid addiction targeting MORs were also presented.
Collapse
Affiliation(s)
- Jia‐Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Chang‐Geng Song
- Department of NeurologyXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ji‐Min Dai
- Department of Hepatobiliary SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ling Li
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiang‐Min Yang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
17
|
Legaki E, Tsaklakidou D, Hatzimanolis A, Segredou E, Petalotis M, Moularogiorgou G, Mouchtouri V, Lykouras L, Stefanis NC, Gazouli M. Association of Alcohol Use Disorder Risk With ADH1B, DRD2, FAAH, SLC39A8, GCKR, and PDYN Genetic Polymorphisms. In Vivo 2022; 36:2092-2104. [PMID: 36099111 PMCID: PMC9463892 DOI: 10.21873/invivo.12935] [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: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Alcohol use disorder (AUD) is a chronic, multifactorial psychiatric condition with an enormous impact on public health and social cost. Genetic studies suggest a heritability, and genome-wide association studies (GWAS) have revealed genetic polymorphisms influencing AUD development. Our study aimed to investigate known variants located in ADH1B, DRD2, FAAH, SLC39A8, GCKR, and PDYN genes (rs1229984, rs7121986, rs324420, rs13107325, rs1260326, rs2281285 respectively) in an AUD Greek cohort in order to shed more light on the genetic predisposition to AUD. MATERIALS AND METHODS Alcohol-dependent individuals (n=251) meeting both the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) and the ICD-10 guidelines for alcohol abuse and dependence, and control individuals (n=280) were recruited. DNA was extracted from whole blood and PCR-restriction fragment length polymorphism (RFLP-PCR) or allele-specific PCR method was used for genotyping. RESULTS Individuals carrying the FAAH rs324420 A allele were significantly associated with increased risk of AUD (p<0.0001). SLC39A8 rs13107325 T allele and ADH1B rs1229984 T allele are overrepresented in control subjects (p<0.0001 and p<0.0001, respectively). The associations are maintained following an adjustment for age and sex and Bonferroni correction. GCKR rs13107325, DRD2 rs7121986, and PDYN rs2281285 polymorphisms did not show a significant association with AUD in the studied population after Bonferroni correction. CONCLUSION Susceptibility to AUD is related to variations in FAAH, ADH1B, and SLC39A8 genes. These polymorphisms could serve as potential biomarkers for AUD risk.
Collapse
Affiliation(s)
- Evangelia Legaki
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Domna Tsaklakidou
- Addiction Unit, 2nd University Department of Psychiatry, "Attikon" University General Hospital, Athens, Greece
| | - Alex Hatzimanolis
- First Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Eirini Segredou
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | - Markos Petalotis
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | | | - Varvara Mouchtouri
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | - Lefteris Lykouras
- Addiction Unit, 2nd University Department of Psychiatry, "Attikon" University General Hospital, Athens, Greece
| | - Nikos C Stefanis
- First Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece;
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece;
- 2 Department of Radiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Sciences, Hellenic Open University, Patras, Greece
| |
Collapse
|
18
|
Non-coding RNA in alcohol use disorder by affecting synaptic plasticity. Exp Brain Res 2022; 240:365-379. [PMID: 35028694 DOI: 10.1007/s00221-022-06305-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD) is one of the most serious public health problems worldwide. AUD is a complex disorder, and there is ample evidence that genetic predisposition is critical to its development. Recent studies have shown that genetic predisposition leads to the onset of AUD, and alcohol metabolism can affect epigenetic inheritance, which in turn affects synaptic plasticity, alters brain function, and leads to more severe addictive behaviors. Non-coding RNAs (ncRNAs), especially microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play an important role in alcohol addiction. This paper reviews the regulatory role of ncRNAs. ncRNAs are involved in enzyme and neurotransmitter reaction systems during alcohol use disorder. Alcohol consumption regulates the expression of ncRNAs that mediate epigenetic modification and synaptic plasticity, which play an important role in the development of chronic AUD. ncRNAs may be used not only as predictors of therapeutic responses but also as therapeutic targets of AUD. Chronic alcoholism is more likely to lead to neuroimmune disorders, including permanent brain dysfunction. AUD induced by long-term alcoholism greatly alters the expression of genes in the human genome, especially the expression of ncRNAs. Alcohol can cause a series of pathological changes by interfering with gene expression, such as through disordered miRNA-mRNA expression networks, epigenetic modifications, disordered metabolism, and even synaptic remodeling. ncRNAs are involved in the transition from moderate drinking to alcohol dependence.
Collapse
|
19
|
de Zavalia N, Schoettner K, Goldsmith JA, Solis P, Ferraro S, Parent G, Amir S. Bmal1 in the striatum influences alcohol intake in a sexually dimorphic manner. Commun Biol 2021; 4:1227. [PMID: 34702951 PMCID: PMC8548330 DOI: 10.1038/s42003-021-02715-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023] Open
Abstract
Alcohol consumption has been strongly associated with circadian clock gene expression in mammals. Analysis of clock genes revealed a potential role of Bmal1 in the control of alcohol drinking behavior. However, a causal role of Bmal1 and neural pathways through which it may influence alcohol intake have not yet been established. Here we show that selective ablation of Bmal1 (Cre/loxP system) from medium spiny neurons of the striatum induces sexual dimorphic alterations in alcohol consumption in mice, resulting in augmentation of voluntary alcohol intake in males and repression of intake in females. Per2mRNA expression, quantified by qPCR, decreases in the striatum after the deletion of Bmal1. To address the possibility that the effect of striatal Bmal1 deletion on alcohol intake and preference involves changes in the local expression of Per2, voluntary alcohol intake (two-bottle, free-choice paradigm) was studied in mice with a selective ablation of Per2 from medium spiny neurons of the striatum. Striatal ablation of Per2 increases voluntary alcohol intake in males but has no effect in females. Striatal Bmal1 and Per2 expression thus may contribute to the propensity to consume alcohol in a sex -specific manner in mice.
Collapse
Affiliation(s)
- Nuria de Zavalia
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada.
| | - Konrad Schoettner
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada
| | - Jory A Goldsmith
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada
| | - Pavel Solis
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada
| | - Sarah Ferraro
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada
| | - Gabrielle Parent
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada
| | - Shimon Amir
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Canada.
| |
Collapse
|
20
|
Leclercq S, Le Roy T, Furgiuele S, Coste V, Bindels LB, Leyrolle Q, Neyrinck AM, Quoilin C, Amadieu C, Petit G, Dricot L, Tagliatti V, Cani PD, Verbeke K, Colet JM, Stärkel P, de Timary P, Delzenne NM. Gut Microbiota-Induced Changes in β-Hydroxybutyrate Metabolism Are Linked to Altered Sociability and Depression in Alcohol Use Disorder. Cell Rep 2021; 33:108238. [PMID: 33053357 DOI: 10.1016/j.celrep.2020.108238] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 08/04/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with alcohol use disorder (AUD) present with important emotional, cognitive, and social impairments. The gut microbiota has been recently shown to regulate brain functions and behavior but convincing evidence of its role in AUD is lacking. Here, we show that gut dysbiosis is associated with metabolic alterations that affect behavioral (depression, sociability) and neurobiological (myelination, neurotransmission, inflammation) processes involved in alcohol addiction. By transplanting the gut microbiota from AUD patients to mice, we point out that the production of ethanol by specific bacterial genera and the reduction of lipolysis are associated with a lower hepatic synthesis of β-hydroxybutyrate (BHB), which thereby prevents the neuroprotective effect of BHB. We confirm these results in detoxified AUD patients, in which we observe a persisting ethanol production in the feces as well as correlations among low plasma BHB levels and social impairments, depression, or brain white matter alterations.
Collapse
Affiliation(s)
- Sophie Leclercq
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Tiphaine Le Roy
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Sonia Furgiuele
- Laboratory of Human Biology & Toxicology, UMONS, 7000 Mons, Belgium
| | - Valentin Coste
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Quentin Leyrolle
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Caroline Quoilin
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Camille Amadieu
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Géraldine Petit
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laurence Dricot
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | | | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Kristin Verbeke
- Translational Research Center in Gastrointestinal Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Jean-Marie Colet
- Laboratory of Human Biology & Toxicology, UMONS, 7000 Mons, Belgium
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Department of Hepatogastroenterology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Philippe de Timary
- Institute of Neuroscience, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium.
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
21
|
Li L, Yu H, Liu Y, Meng YJ, Li XJ, Zhang C, Liang S, Li ML, Guo W, QiangWang, Deng W, Ma X, Coid J, Li T. Lower regional grey matter in alcohol use disorders: evidence from a voxel-based meta-analysis. BMC Psychiatry 2021; 21:247. [PMID: 33975595 PMCID: PMC8111920 DOI: 10.1186/s12888-021-03244-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/28/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Previous research using whole-brain neuroimaging techniques has revealed structural differences of grey matter (GM) in alcohol use disorder (AUD) patients. However, some of the findings diverge from other neuroimaging studies and require further replication. The quantity of relevant research has, thus far, been limited and the association between GM and abstinence duration of AUD patients has not yet been systematically reviewed. METHODS The present research conducted a meta-analysis of voxel-based GM studies in AUD patients published before Jan 2021. The study utilised a whole brain-based d-mapping approach to explore GM changes in AUD patients, and further analysed the relationship between GM deficits, abstinence duration and individual differences. RESULTS The current research included 23 studies with a sample size of 846 AUD patients and 878 controls. The d-mapping approach identified lower GM in brain regions including the right cingulate gyrus, right insula and left middle frontal gyrus in AUD patients compared to controls. Meta-regression analyses found increasing GM atrophy in the right insula associated with the longer mean abstinence duration of the samples in the studies in our analysis. GM atrophy was also found positively correlated with the mean age of the samples in the right insula, and positively correlated with male ratio in the left middle frontal gyrus. CONCLUSIONS GM atrophy was found in the cingulate gyrus and insula in AUD patients. These findings align with published meta-analyses, suggesting they are potential deficits for AUD patients. Abstinence duration, age and gender also affect GM atrophy in AUD patients. This research provides some evidence of the underlying neuroanatomical nature of AUD.
Collapse
Affiliation(s)
- Lei Li
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Hua Yu
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yihao Liu
- grid.8391.30000 0004 1936 8024Department of Psychology, College of Life and Environmental Science, University of Exeter, Exeter, UK
| | - Ya-jing Meng
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao-jing Li
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chengcheng Zhang
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Sugai Liang
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Ming-li Li
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Wanjun Guo
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - QiangWang
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Deng
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaohong Ma
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jeremy Coid
- grid.412901.f0000 0004 1770 1022Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.13291.380000 0001 0807 1581Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.412901.f0000 0004 1770 1022Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Li
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China. .,Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China. .,Brain Research Center, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
Effect of chronic alcohol intake on motor functions on the elderly. Neurosci Lett 2021; 745:135630. [PMID: 33440234 DOI: 10.1016/j.neulet.2021.135630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 12/21/2022]
Abstract
Alcohol use disorder (AUD) is a chronic and progressive disease influenced by genetic, psychosocial, and environmental factors. The consequences of alcohol consumption involve alterations in neural circuits of emotion and cognition, as well as in the motor planning circuit. Furthermore, during the natural aging process, several biochemical and functional alterations are also observed with neurological consequences. Thus, considering the consequences of chronic alcohol consumption on neural systems and natural aging process, we aimed to analyze the degree of motor and functional impairment in elderly with chronic alcohol consumption. Sixty elderly underwent an analysis of alcohol consumption profile (Alcohol Use Disorders Identification Test - AUDIT) that divided them into a control group (CON) and an alcohol group (ALC). The analysis of quality of life was performed using the Medical Outcomes Study 36-Item Short-Form Health Survey (SF-36), the analysis of motor function was performed using the Borg Scale, the Six-Minute Walk Test (6MWT) and the Motor Scale for Elderly (MSE). We were able to conclude that the misuse of alcohol by the elderly promotes significant physical limitations. These limitations result in a worsening of functional capacity of walking and various dimensions of motor ability: fine motor skill, global coordination, balance, body scheme, spatial organization, temporal organization, and general motor aptitude. Besides the physical limitations caused by alcohol use, the quality of life in their physical, mental, and social aspects was reduced. Thus, actions are required to help the elderly understand these losses and exercise control over alcohol misuse.
Collapse
|
23
|
Zhang D, Dong X, Liu X, Ye L, Li S, Zhu R, Ye Y, Jiang Y. Proteomic Analysis of Brain Regions Reveals Brain Regional Differences and the Involvement of Multiple Keratins in Chronic Alcohol Neurotoxicity. Alcohol Alcohol 2020; 55:147-156. [PMID: 32047899 DOI: 10.1093/alcalc/agaa007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/22/2019] [Accepted: 01/13/2020] [Indexed: 12/29/2022] Open
Abstract
AIMS Alcohol abuse has attracted public attention and chronic alcohol exposure can result in irreversible structural changes in the brain. The molecular mechanisms underlying alcohol neurotoxicity are complex, mandating comprehensive mining of spatial protein expression profile. METHODS In this study, mice models of chronic alcohol intoxication were established after 95% alcohol vapor administration for 30 consecutive days. On Day 30, striatum (the dorsal and ventral striatum) and hippocampus, the two major brain regions responsible for learning and memorizing while being sensitive to alcohol toxicity, were collected. After that, isobaric tags for relative and absolute quantitation -based quantitative proteomic analysis were carried out for further exploration of the novel mechanisms underlying alcohol neurotoxicity. RESULTS Proteomic results showed that in the striatum, 29 proteins were significantly up-regulated and 17 proteins were significantly down-regulated. In the hippocampus, 72 proteins were significantly up-regulated, while 2 proteins were significantly down-regulated. Analysis of the overlay proteins revealed that a total of 102 proteins were consistently altered (P < 0.05) in both hippocampus and striatum regions, including multiple keratins such as Krt6a, Krt17 and Krt5. Ingenuity pathway analysis revealed that previously reported diseases/biofunctions such as dermatological diseases and developmental disorders were enriched in those proteins. Interestingly, the glucocorticoid receptor (GR) signaling was among the top enriched pathways in both brain regions, while multiple keratins from the GR signaling such as Krt1 and Krt17 exhibited significantly opposite expression patterns in the two brain nuclei. Moreover, there are several other involved pathways significantly differed between the hippocampus and striatum. CONCLUSIONS Our data revealed brain regional differences upon alcohol consumption and indicated the critical involvement of keratins from GR signaling in alcohol neurotoxicity. The differences in proteomic results between the striatum and hippocampus suggested a necessity of taking into consideration brain regional differences and intertwined signaling pathways rather than merely focusing on single nuclei or molecule during the study of drug-induced neurotoxicity in the future.
Collapse
Affiliation(s)
- Dingang Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoru Dong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaochen Liu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lin Ye
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shuhao Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Rongzhe Zhu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yonghong Ye
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
24
|
Brain anatomical covariation patterns linked to binge drinking and age at first full drink. NEUROIMAGE-CLINICAL 2020; 29:102529. [PMID: 33321271 PMCID: PMC7745054 DOI: 10.1016/j.nicl.2020.102529] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/10/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022]
Abstract
We identified a reproducible cortical and subcortical brain structural covariation pattern. A novel pattern discovery method Joint and Individual Variance Explained (JIVE) was used. The cortical and subcortical structural covariation pattern is related to alcohol use initiation. The identified pattern is dominated by covariation among brainstem, thalamus and PFC. A thalamic-PFC-brainstem circuitry might be related to alcohol use initiation.
Binge drinking and age at first full drink (AFD) of alcohol prior to 21 years (AFD < 21) have been linked to neuroanatomical differences in cortical and subcortical grey matter (GM) volume, cortical thickness, and surface area. Despite the importance of understanding network-level relationships, structural covariation patterns among these morphological measures have yet to be examined in relation to binge drinking and AFD < 21. Here, we used the Joint and Individual Variance Explained (JIVE) method to characterize structural covariation patterns common across and specific to morphological measures in 293 participants (149 individuals with past-12-month binge drinking and 144 healthy controls) from the Human Connectome Project (HCP). An independent dataset (Nathan Kline Institute Rockland Sample; NKI-RS) was used to examine reproducibility/generalizability. We identified a reproducible joint component dominated by structural covariation between GM volume in the brainstem and thalamus proper, and GM volume and surface area in prefrontal cortical regions. Using linear mixed regression models, we found that participants with AFD < 21 showed lower joint component scores in both the HCP (beta = 0.059, p-value = 0.016; Cohen’s d = 0.441) and NKI-RS (beta = 0.023, p-value = 0.040, Cohen’s d = 0.216) datasets, whereas the individual thickness component associated with binge drinking (p-value = 0.02) and AFD < 21 (p-value < 0.001) in the HCP dataset was not statistically significant in the NKI-RS sample. Our findings were also generalizable to the HCP full sample (n = 880 participants). Taken together, our results show that use of JIVE analysis in high-dimensional, large-scale, psychiatry-related datasets led to discovery of a reproducible cortical and subcortical structural covariation pattern involving brain regions relevant to thalamic-PFC-brainstem neural circuitry which is related to AFD < 21 and suggests a possible extension of existing addiction neurocircuitry in humans.
Collapse
|
25
|
Acute Alcohol Effects on Response Inhibition Depend on Response Automatization, but not on GABA or Glutamate Levels in the ACC and Striatum. J Clin Med 2020; 9:jcm9020481. [PMID: 32050509 PMCID: PMC7073826 DOI: 10.3390/jcm9020481] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Alcohol increases GABAergic signaling and decreases glutamatergic signaling in the brain. Variations in these neurotransmitter levels may modulate/predict executive functioning. Matching this, strong impairments of response inhibition are one of the most consistently reported cognitive/behavioral effects of acute alcohol intoxication. However, it has never been investigated whether baseline differences in these neurotransmitters allow to predict how much alcohol intoxication impairs response inhibition, and whether this is reflected in neurophysiological measures of cognitive control. We used MR spectroscopy to assess baseline (i.e., sober) GABA and glutamate levels in the anterior cingulate cortex (ACC) and striatum in n = 30 healthy young males, who were subsequently tested once sober and once intoxicated (1.01 permille). Inhibition was assessed with the sustained attention to response task (SART). This paradigm also allows to examine the effect of different degrees of response automatization, which is a known modulator for response inhibition, but does not seem to be substantially impaired during acute intoxication. As a neurophysiological correlate of response inhibition and control, we quantified EEG-derived theta band power and located its source using beamforming analyses. We found that alcohol-induced response inhibition deficits only occurred in the case of response automatization. This was reflected by decreased theta band activity in the left supplementary motor area (SMA), which may reflect modulations in the encoding of a surprise signal in response to inhibition cues. However, we did not find that differences in baseline (i.e., sober) GABA or glutamate levels significantly modulated differences in the size of alcohol-induced inhibition deficits.
Collapse
|
26
|
Kim JS, Martin-Fardon R. Possible Role of CRF-Hcrt Interaction in the Infralimbic Cortex in the Emergence and Maintenance of Compulsive Alcohol-Seeking Behavior. Alcohol Clin Exp Res 2020; 44:354-367. [PMID: 31840823 PMCID: PMC7018591 DOI: 10.1111/acer.14264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder that is characterized by the compulsive use of alcohol despite numerous health, social, and economic consequences. Initially, the use of alcohol is driven by positive reinforcement. Over time, however, alcohol use can take on a compulsive quality that is driven by the desire to avoid the negative consequences of abstinence, including negative affect and heightened stress/anxiety. This transition from positive reinforcement- to negative reinforcement-driven consumption involves the corticotropin-releasing factor (CRF) system, although mounting evidence now suggests that the CRF system interacts with other neural systems to ultimately produce behaviors that are symptomatic of compulsive alcohol use, such as the hypocretin (Hcrt) system. Hypocretins are produced exclusively in the hypothalamus, but Hcrt neurons project widely throughout the brain and reach regions that perform regulatory functions for numerous behavioral and physiological responses-including the infralimbic cortex (IL) of the medial prefrontal cortex (mPFC). Although the entire mPFC undergoes neuroadaptive changes following prolonged alcohol exposure, the IL appears to undergo more robust changes compared with other mPFC substructures. Evidence to date suggests that the IL is likely involved in EtOH-seeking behavior, but ambiguities with respect to the specific role of the IL in this regard make it difficult to draw definitive conclusions. Furthermore, the manner in which CRF interacts with Hcrt in this region as it pertains to alcohol-seeking behavior is largely unknown, although immunohistochemical and electrophysiological experiments have shown that CRF and Hcrt directly interact in the mPFC, suggesting that the interaction between CRF and Hcrt in the IL may be critically important for the development and subsequent maintenance of compulsive alcohol seeking. This review aims to consolidate recent literature regarding the role of the IL in alcohol-seeking behavior and to discuss evidence that supports a functional interaction between Hcrt and CRF in the IL.
Collapse
Affiliation(s)
- Jung S. Kim
- Department of Molecular Medicine, Scripps Research, La Jolla, USA
| | | |
Collapse
|
27
|
Bryant VE, Gullett JM, Porges EC, Cook RL, Bryant KJ, Woods AJ, Williamson J, Ennis N, Cohen RA. History of Alcohol Consumption and HIV Status Related to Functional Connectivity Differences in the Brain During Working Memory Performance. Curr HIV Res 2020; 18:181-193. [PMID: 32065091 PMCID: PMC7315564 DOI: 10.2174/1570162x18666200217100123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/25/2020] [Accepted: 02/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Poorer working memory function has previously been associated with alcohol misuse, Human Immunodeficiency Virus (HIV) positive status, and risky behavior. Poorer working memory performance relates to alterations in specific brain networks. OBJECTIVE The current study examined if there was a relationship between brain networks involved in working memory and reported level of alcohol consumption during an individual's period of heaviest use. Furthermore, we examined whether HIV status and the interaction between HIV and alcohol consumption was associated with differences in these brain networks. METHODS Fifty adults, 26 of whom were HIV positive, engaged in an n-back working memory task (0-back and 2-back trials) administered in a magnetic resonance imaging (MRI) scanner. The Kreek- McHugh-Schluger-Kellogg (KMSK) scale of alcohol consumption was used to characterize an individual's period of heaviest use and correlates well with their risk for alcohol dependence. Connectivity analyses were conducted using data collected during n-back task. RESULTS Functional connectivity differences associated with greater alcohol consumption included negative connectivity, primarily from parietal attention networks to frontal networks. Greater alcohol consumption was also associated with positive connectivity from working memory nodes to the precuneus and paracingulate. HIV positive status was associated with more nodes of negative functional connectivity relative to alcohol consumption history alone, particularly in the frontoparietal networks. The HIV positive individuals with heavier drinking history related to negative fronto-parietal connectivity, along with positive connectivity from working memory nodes to mesolimbic regions. CONCLUSION Findings allow for a better understanding of brain networks affected by HIV and alcohol and may provide avenues for interventions.
Collapse
Affiliation(s)
- Vaughn E Bryant
- Department of Epidemiology, Southern HIV and Alcohol Research Consortium and Center, University of Florida, Gainesville, FL, United States
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Joseph M Gullett
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Eric C Porges
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Robert L Cook
- Department of Epidemiology, Southern HIV and Alcohol Research Consortium and Center, University of Florida, Gainesville, FL, United States
| | - Kendall J Bryant
- National Institute of Alcohol Abuse and Alcoholism, MD, United States
| | - Adam J Woods
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - John Williamson
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Nicole Ennis
- Department of Behavioral Sciences and Social Medicine, Florida State University, Tallahassee, FL, United States
| | - Ronald A Cohen
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
28
|
Evangelou E, Gao H, Chu C, Ntritsos G, Blakeley P, Butts AR, Pazoki R, Suzuki H, Koskeridis F, Yiorkas AM, Karaman I, Elliott J, Luo Q, Aeschbacher S, Bartz TM, Baumeister SE, Braund PS, Brown MR, Brody JA, Clarke TK, Dimou N, Faul JD, Homuth G, Jackson AU, Kentistou KA, Joshi PK, Lemaitre RN, Lind PA, Lyytikäinen LP, Mangino M, Milaneschi Y, Nelson CP, Nolte IM, Perälä MM, Polasek O, Porteous D, Ratliff SM, Smith JA, Stančáková A, Teumer A, Tuominen S, Thériault S, Vangipurapu J, Whitfield JB, Wood A, Yao J, Yu B, Zhao W, Arking DE, Auvinen J, Liu C, Männikkö M, Risch L, Rotter JI, Snieder H, Veijola J, Blakemore AI, Boehnke M, Campbell H, Conen D, Eriksson JG, Grabe HJ, Guo X, van der Harst P, Hartman CA, Hayward C, Heath AC, Jarvelin MR, Kähönen M, Kardia SLR, Kühne M, Kuusisto J, Laakso M, Lahti J, Lehtimäki T, McIntosh AM, Mohlke KL, Morrison AC, Martin NG, Oldehinkel AJ, Penninx BWJH, Psaty BM, Raitakari OT, Rudan I, Samani NJ, Scott LJ, Spector TD, Verweij N, Weir DR, Wilson JF, Levy D, Tzoulaki I, Bell JD, Matthews PM, Rothenfluh A, Desrivières S, Schumann G, Elliott P. New alcohol-related genes suggest shared genetic mechanisms with neuropsychiatric disorders. Nat Hum Behav 2019; 3:950-961. [PMID: 31358974 PMCID: PMC7711277 DOI: 10.1038/s41562-019-0653-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Excessive alcohol consumption is one of the main causes of death and disability worldwide. Alcohol consumption is a heritable complex trait. Here we conducted a meta-analysis of genome-wide association studies of alcohol consumption (g d-1) from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake. We identified 46 new common loci and investigated their potential functional importance using magnetic resonance imaging data and gene expression studies. We identify genetic pathways associated with alcohol consumption and suggest genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - He Gao
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Congying Chu
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Paul Blakeley
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, ITMAT Data Science Group, Imperial College London, London, UK
| | - Andrew R Butts
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Raha Pazoki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Hideaki Suzuki
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Andrianos M Yiorkas
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Ibrahim Karaman
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Joshua Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Department of Psychology and the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | | | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sebastian E Baumeister
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Chair of Epidemiology, Ludwig-Maximilians-Universitat Munchen, UNIKA-T Augsburg, Augsburg, Germany
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Toni-Kim Clarke
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Niki Dimou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A Kentistou
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas Foundation Trust, London, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mia-Maria Perälä
- Folkhälsan Research Center, Helsinki, Finland
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - David Porteous
- Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Scott M Ratliff
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Samuli Tuominen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alexis Wood
- Department of Pediatrics/Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juha Auvinen
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Oulunkaari Health Center, Ii, Finland
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Minna Männikkö
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Lorenz Risch
- Institute of Clinical Chemistry, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Labormedizinisches Zentrum Dr. Risch, Vaduz, Liechtenstein
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
- Department of Psychiatry, University Hospital of Oulu, Oulu, Finland
- Medical research Center Oulu, University and University Hospital of Oulu, Oulu, Finland
| | - Alexandra I Blakemore
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Catharina A Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Andrew C Heath
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Michael Kühne
- Cardiology Division, University Hospital Basel, Basel, Switzerland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
| | - Andrew M McIntosh
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Albertine J Oldehinkel
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | - Paul M Matthews
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Adrian Rothenfluh
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Departments of Psychiatry, Neurobiology & Anatomy, Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- PONS Research Group, Dept of Psychiatry and Psychotherapy, Campus Charite Mitte, Humboldt University, Berlin, Germany and Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, P.R. China.
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK.
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College Healthcare NHS Trust and Imperial College London, London, UK.
- Health Data Research UK London Substantive Site, London, UK.
| |
Collapse
|
29
|
Increased Ethanol Consumption and Locomotion Develop upon Ethanol Deprivation in Rats Overexpressing the Adenosine (A) 2A Receptor. Neuroscience 2019; 418:133-148. [PMID: 31449988 DOI: 10.1016/j.neuroscience.2019.08.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/04/2019] [Accepted: 08/17/2019] [Indexed: 02/03/2023]
Abstract
Preclinical data indicate that ethanol produces behavioral effects that can be regulated by many neurotransmitters and neuromodulators like adenosine (A). The most important receptors with respect to the rewarding effects of ethanol seem to be the A2A receptors. This study used a transgenic strategy, specifically rats overexpressing the A2A receptor, to characterize the neurobiological mechanisms of ethanol consumption as measured by intermittent access to 20% ethanol in a two-bottle choice paradigm. In this model, no change in ethanol consumption was observed in transgenic animals compared to wild type controls during the acquisition/maintenance phase. Following alcohol deprivation, only transgenic rats overexpressing the A2A receptor exhibited escalation of ethanol consumption and drank more (by ca. 90%), but not significantly, ethanol than did the wild type rats. During ethanol withdrawal, the immobility time of rats overexpressing the A2A receptor in the forced swim test was lower than that of wild type rats. Moreover, transgenic rats withdrawn from ethanol, compared to the drug-naive transgenic animals, exhibited an increase above 70% in locomotion. The results indicated that the overexpression of A2A receptors may be a risk factor for the escalation of ethanol consumption despite the reduction in depression-like signs of ethanol withdrawal.
Collapse
|
30
|
Nonphosphorylatable Src Ser75 Mutation Increases Ethanol Preference and Consumption in Mice. eNeuro 2019; 6:eN-NWR-0418-18. [PMID: 30963106 PMCID: PMC6451160 DOI: 10.1523/eneuro.0418-18.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 11/21/2022] Open
Abstract
Src is highly expressed in CNS neurons and contributes not only to developmental proliferation and differentiation but also to high-order brain functions, such as those contributing to alcohol consumption. Src knock-out mice exhibit no CNS abnormalities, presumably due to compensation by other Src family kinases (SFKs), but have a shortened lifespan and osteopetrosis-associated defects, impeding investigations of the role of Src on behavior in adult mice. However, the Unique domain of Src differs from those in other SFKs and is phosphorylated by cyclin-dependent kinase 1 (Cdk1) and Cdk5 at Ser75, which influences its postmitotic function in neurons. Therefore, ethanol consumption in mice harboring nonphosphorylatable (Ser75Ala) or phosphomimetic (Ser75Asp) Src mutants was investigated. Mice harboring the Ser75Ala Src mutant, but not the Ser75Asp mutant, had a higher preference for and consumption of solutions containing 5% and 10% ethanol than wild-type mice. However, plasma ethanol concentrations and sensitivities to the sedative effects of ethanol were not different among the groups. In mice harboring the Ser75Ala Src mutant, the activity of Rho-associated kinase (ROCK) in the striatum was significantly lower and Akt Ser473 phosphorylation was significantly higher than in wild-type mice. These results suggest that Src regulates voluntary ethanol drinking in a manner that depends on Ser75 phosphorylation.
Collapse
|
31
|
Wang WT, Lee P, Hui D, Michaelis EK, Choi IY. Effects of Ethanol Exposure on the Neurochemical Profile of a Transgenic Mouse Model with Enhanced Glutamate Release Using In Vivo 1H MRS. Neurochem Res 2019; 44:133-146. [PMID: 30334175 PMCID: PMC6497580 DOI: 10.1007/s11064-018-2658-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022]
Abstract
Ethanol (EtOH) intake leads to modulation of glutamatergic transmission, which may contribute to ethanol intoxication, tolerance and dependence. To study metabolic responses to the hyper glutamatergic status at synapses during ethanol exposure, we used Glud1 transgenic (tg) mice that over-express the enzyme glutamate dehydrogenase in brain neurons and release excess glutamate (Glu) in synapses. We measured neurochemical changes in the hippocampus and striatum of tg and wild-type (wt) mice using proton magnetic resonance spectroscopy before and after the animals were fed with diets within which EtOH constituting up to 6.4% of total calories for 24 weeks. In the hippocampus, the EtOH diet led to significant increases in concentrations of EtOH, glutamine (Gln), Glu, phosphocholine (PCho), taurine, and Gln + Glu, when compared with their baseline concentrations. In the striatum, the EtOH diet led to significant increases in concentrations of GABA, Gln, Gln + Glu, and PCho. In general, neurochemical changes were more pronounced in the striatum than the hippocampus in both tg and wt mice. Overall neurochemical changes due to EtOH exposure were very similar in tg and wt mice. This study describes time courses of neurochemical profiles before and during chronic EtOH exposure, which can serve as a reference for future studies investigating ethanol-induced neurochemical changes.
Collapse
Affiliation(s)
- Wen-Tung Wang
- Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Phil Lee
- Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dongwei Hui
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Elias K Michaelis
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - In-Young Choi
- Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
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
|
Ulenius L, Adermark L, Söderpalm B, Ericson M. Ethanol-Induced Taurine Elevation in the Rat Dorsal Striatum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:173-181. [PMID: 28849453 DOI: 10.1007/978-94-024-1079-2_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
In the search for the primary mechanism underlying the dopamine elevating properties of ethanol we have established that raised levels of taurine in the nucleus accumbens (nAc) is pivotal. In the nAc, the release of taurine appears to be connected to osmoregulation, and neither taurine nor dopamine is increased if ethanol is administered in a hypertonic saline solution. However, even though the nAc is important for drug-reinforcement, manifestation of addiction has been postulated to recruit the more dorsal parts of the striatum (DS). How ethanol influences dopamine and taurine in the DS and their role in addiction is thus far poorly understood. By means of in vivo microdialysis in freely moving rats we concomitantly monitored extracellular levels of dopamine and taurine in the DS following administration of ethanol diluted either in an isotonic or hypertonic saline solution. In a different set of rats, placed in a voluntary ethanol consumption paradigm (intermittent access to 20% ethanol for 2 months), taurine and dopamine were monitored following an acute injection of ethanol. We found that neither administration of ethanol diluted in a hypertonic saline solution, nor 2 months of moderate ethanol consumption, influence the ethanol-induced increase of taurine in the DS. We propose that there may be regional differences in the relationship between taurine, dopamine and ethanol in the nAc and in the DS. It remains to be determined if this subregion-specificity is important for the transition from recreational drug use to a compulsive habit.
Collapse
Affiliation(s)
- Lisa Ulenius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Czoty PW, John WS, Newman AH, Nader MA. Yawning elicited by intravenous ethanol in rhesus monkeys with experience self-administering cocaine and ethanol: Involvement of dopamine D 3 receptors. Alcohol 2018; 69:1-5. [PMID: 29550583 PMCID: PMC5904012 DOI: 10.1016/j.alcohol.2017.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/27/2022]
Abstract
Characterization of the effects of long-term alcohol consumption on the brain would be aided by the development of behavioral assays that are relatively easy to implement in animal models of alcohol use disorders. Assessing unconditioned behaviors, such as drug-elicited yawning in models that permit long-term alcohol ingestion, may be a valuable complement to more invasive and costly procedures. The present studies investigated previous unexpected findings of ethanol-induced yawning in nonhuman primates. Subjects were adult male rhesus monkeys (n = 8), all of which had experience self-administering intravenous cocaine for several years. Four monkeys also had experience consuming 2.0 g/kg ethanol over 1 h per day, 5 days per week, for 6.8-12.0 months. All monkeys received saline or ethanol (0.25-1.0 g/kg) infused intravenously (i.v.) over 10 min, and the number of yawns elicited during the infusion was counted. A second experiment in the ethanol-experienced monkeys examined whether ethanol-induced yawning could be blocked by PG01037 (1.0, 3.0 mg/kg, i.v.), a selective antagonist at dopamine D3 receptors (D3R). Ethanol significantly and dose-dependently increased yawns in the ethanol-experienced animals, but not the ethanol-naïve animals. In the ethanol-experienced monkeys, this effect of ethanol was blocked by the D3R antagonist. The pharmacology of yawning is complex and a good deal of model development remains to be performed to characterize the potential involvement of other neurotransmitter systems. Nonetheless, drug-elicited yawning may be a useful unconditioned behavioral assay to assess the effects of long-term alcohol consumption in established nonhuman primate models.
Collapse
Affiliation(s)
- Paul W Czoty
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States.
| | - William S John
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|
35
|
Ruggeri B, Macare C, Stopponi S, Jia T, Carvalho FM, Robert G, Banaschewski T, Bokde ALW, Bromberg U, Büchel C, Cattrell A, Conrod PJ, Desrivières S, Flor H, Frouin V, Gallinat J, Garavan H, Gowland P, Heinz A, Ittermann B, Martinot JL, Paillère Martinot ML, Nees F, Papadopoulos-Orfanos D, Paus T, Poustka L, Smolka MN, Vetter NC, Walter H, Whelan R, Sommer WH, Bakalkin G, Ciccocioppo R, Schumann G. Methylation of OPRL1 mediates the effect of psychosocial stress on binge drinking in adolescents. J Child Psychol Psychiatry 2018; 59:650-658. [PMID: 29197086 PMCID: PMC5975104 DOI: 10.1111/jcpp.12843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nociceptin is a key regulator linking environmental stress and alcohol drinking. In a genome-wide methylation analysis, we recently identified an association of a methylated region in the OPRL1 gene with alcohol-use disorders. METHODS Here, we investigate the biological basis of this observation by analysing psychosocial stressors, methylation of the OPRL1 gene, brain response during reward anticipation and alcohol drinking in 660 fourteen-year-old adolescents of the IMAGEN study. We validate our findings in marchigian sardinian (msP) alcohol-preferring rats that are genetically selected for increased alcohol drinking and stress sensitivity. RESULTS We found that low methylation levels in intron 1 of OPRL1 are associated with higher psychosocial stress and higher frequency of binge drinking, an effect mediated by OPRL1 methylation. In individuals with low methylation of OPRL1, frequency of binge drinking is associated with stronger BOLD response in the ventral striatum during reward anticipation. In msP rats, we found that stress results in increased alcohol intake and decreased methylation of OPRL1 in the nucleus accumbens. CONCLUSIONS Our findings describe an epigenetic mechanism that helps to explain how psychosocial stress influences risky alcohol consumption and reward processing, thus contributing to the elucidation of biological mechanisms underlying risk for substance abuse.
Collapse
Affiliation(s)
- Barbara Ruggeri
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Christine Macare
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | | | - Tianye Jia
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Fabiana M. Carvalho
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Gabriel Robert
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Tobias Banaschewski
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Arun LW Bokde
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Uli Bromberg
- Universitaetsklinikum Hamburg Eppendorf, Hamburg, Germany
| | | | - Anna Cattrell
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Patricia J Conrod
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,Department of Psychiatry, Université de Montreal, Canada
| | - Sylvane Desrivières
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Vincent Frouin
- Neurospin, Commissariat à l'Energie Atomique et aux Energies Alternatives, Paris, France
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Germany
| | - Hugh Garavan
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland,Departments of Psychiatry and Psychology, University of Vermont, USA
| | | | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Germany
| | - Jean Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM CEA Unit 1000 “Imaging & Psychiatry”, University Paris Sud, Orsay
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM CEA Unit 1000 “Imaging & Psychiatry”, University Paris Sud, Orsay
| | - Frauke Nees
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | | | - Tomáš Paus
- School of Psychology, University of Nottingham, UK,Rotman Research Institute, University of Toronto, Toronto, Canada,Child Mind Institute, New York, USA
| | - Luise Poustka
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Germany
| | - Nora C. Vetter
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Germany
| | | | - Wolfgang H Sommer
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Georgy Bakalkin
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - Gunter Schumann
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, UK,MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | | |
Collapse
|
36
|
Blackwell KT, Salinas AG, Tewatia P, English B, Hellgren Kotaleski J, Lovinger DM. Molecular mechanisms underlying striatal synaptic plasticity: relevance to chronic alcohol consumption and seeking. Eur J Neurosci 2018; 49:768-783. [PMID: 29602186 DOI: 10.1111/ejn.13919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 01/02/2023]
Abstract
The striatum, the input structure of the basal ganglia, is a major site of learning and memory for goal-directed actions and habit formation. Spiny projection neurons of the striatum integrate cortical, thalamic, and nigral inputs to learn associations, with cortico-striatal synaptic plasticity as a learning mechanism. Signaling molecules implicated in synaptic plasticity are altered in alcohol withdrawal, which may contribute to overly strong learning and increased alcohol seeking and consumption. To understand how interactions among signaling molecules produce synaptic plasticity, we implemented a mechanistic model of signaling pathways activated by dopamine D1 receptors, acetylcholine receptors, and glutamate. We use our novel, computationally efficient simulator, NeuroRD, to simulate stochastic interactions both within and between dendritic spines. Dopamine release during theta burst and 20-Hz stimulation was extrapolated from fast-scan cyclic voltammetry data collected in mouse striatal slices. Our results show that the combined activity of several key plasticity molecules correctly predicts the occurrence of either LTP, LTD, or no plasticity for numerous experimental protocols. To investigate spatial interactions, we stimulate two spines, either adjacent or separated on a 20-μm dendritic segment. Our results show that molecules underlying LTP exhibit spatial specificity, whereas 2-arachidonoylglycerol exhibits a spatially diffuse elevation. We also implement changes in NMDA receptors, adenylyl cyclase, and G protein signaling that have been measured following chronic alcohol treatment. Simulations under these conditions suggest that the molecular changes can predict changes in synaptic plasticity, thereby accounting for some aspects of alcohol use disorder.
Collapse
Affiliation(s)
- Kim T Blackwell
- The Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA.,Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Armando G Salinas
- The Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA.,National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Parul Tewatia
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Brad English
- The Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Jeanette Hellgren Kotaleski
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David M Lovinger
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
37
|
Cuzon Carlson VC, Grant KA, Lovinger DM. Synaptic adaptations to chronic ethanol intake in male rhesus monkey dorsal striatum depend on age of drinking onset. Neuropharmacology 2018; 131:128-142. [PMID: 29241653 PMCID: PMC5820135 DOI: 10.1016/j.neuropharm.2017.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/20/2017] [Accepted: 12/05/2017] [Indexed: 12/20/2022]
Abstract
One in 12 adults suffer with alcohol use disorder (AUD). Studies suggest the younger the age in which alcohol consumption begins the higher the probability of being diagnosed with AUD. Binge/excessive alcohol drinking involves a transition from flexible to inflexible behavior likely involving the dorsal striatum (caudate and putamen nuclei). A major focus of this study was to examine the effect of age of drinking onset on subsequent chronic, voluntary ethanol intake and dorsal striatal circuitry. Data from rhesus monkeys (n = 45) that started drinking as adolescents, young adults or mature adults confirms an age-related risk for heavy drinking. Striatal neuroadaptations were examined using whole-cell patch clamp electrophysiology to record AMPA receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) and GABAA receptor-mediated miniature inhibitory postsynaptic currents (mIPSCs) from medium-sized spiny projection neurons located in the caudate or putamen nuclei. In controls, greater GABAergic transmission (mIPSC frequency and amplitude) was observed in the putamen compared to the caudate. With advancing age, in the absence of ethanol, an increase in mIPSC frequency concomitant with changes in mIPSC amplitude was observed in both regions. Chronic ethanol drinking decreased mIPSC frequency in the putamen regardless of age of onset. In the caudate, an ethanol drinking-induced increase in mIPSC frequency was only observed in monkeys that began drinking as young adults. Glutamatergic transmission did not differ between the dorsal striatal subregions in controls. With chronic ethanol drinking there was a decrease in the postsynaptic characteristics of rise time and area of mEPSCs in the putamen but an increase in mEPSC frequency in the caudate. Together, the observed changes in striatal physiology indicate a combined disinhibition due to youth and ethanol leading to abnormally strong activation of the putamen that could contribute to the increased risk for problem drinking in younger drinkers.
Collapse
Affiliation(s)
- Verginia C Cuzon Carlson
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, United States; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, United States
| | - David M Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, United States.
| |
Collapse
|
38
|
Goodwani S, Saternos H, Alasmari F, Sari Y. Metabotropic and ionotropic glutamate receptors as potential targets for the treatment of alcohol use disorder. Neurosci Biobehav Rev 2017; 77:14-31. [PMID: 28242339 DOI: 10.1016/j.neubiorev.2017.02.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicates that dysfunctional glutamate neurotransmission is critical in the initiation and development of alcohol and drug dependence. Alcohol consumption induced downregulation of glutamate transporter 1 (GLT-1) as reported in previous studies from our laboratory. Glutamate is the major excitatory neurotransmitter in the brain, which acts via interactions with several glutamate receptors. Alcohol consumption interferes with the glutamatergic signal transmission by altering the functions of these receptors. Among the glutamate receptors involved in alcohol-drinking behavior are the metabotropic receptors such as mGluR1/5, mGluR2/3, and mGluR7, as well as the ionotropic receptors, NMDA and AMPA. Preclinical studies using agonists and antagonists implicate these glutamatergic receptors in the development of alcohol use disorder (AUD). Therefore, the purpose of this review is to discuss the neurocircuitry involving glutamate transmission in animals exposed to alcohol and further outline the role of metabotropic and ionotropic receptors in the regulation of alcohol-drinking behavior. This review provides ample information about the potential therapeutic role of glutamatergic receptors for the treatment of AUD.
Collapse
Affiliation(s)
- Sunil Goodwani
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA; The Neurodegeneration Consortium, Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Hannah Saternos
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA
| | - Fawaz Alasmari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA
| | - Youssef Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, OH 43614, USA.
| |
Collapse
|
39
|
Buck KJ, Chen G, Kozell LB. Limbic circuitry activation in ethanol withdrawal is regulated by a chromosome 1 locus. Alcohol 2017; 58:153-160. [PMID: 27989609 DOI: 10.1016/j.alcohol.2016.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/03/2016] [Accepted: 09/03/2016] [Indexed: 11/19/2022]
Abstract
Physiological dependence and associated withdrawal episodes are thought to constitute a motivational force sustaining alcohol use/abuse and contributing to relapse in alcoholics. Although no animal model exactly duplicates alcoholism, models for specific factors, including the withdrawal syndrome, are useful for identifying potential genetic and neural determinants of liability in humans. We previously identified highly significant quantitative trait loci (QTLs) with large effects on predisposition to withdrawal after chronic and acute alcohol exposure in mice and mapped these loci to the same region of chromosome 1 (Alcdp1 and Alcw1, respectively). The present studies utilize a novel Alcdp1/Alcw1 congenic model (in which an interval spanning Alcdp1 and Alcw1 from the C57BL/6J donor strain [build GRCm38 150.3-174.6 Mb] has been introgressed onto a uniform inbred DBA/2J genetic background) known to demonstrate significantly less severe chronic and acute withdrawal compared to appropriate background strain animals. Here, using c-Fos induction as a high-resolution marker of neuronal activation, we report that male Alcdp1/Alcw1 congenic animals demonstrate significantly less alcohol withdrawal-associated neural activation compared to appropriate background strain animals in the prelimbic and cingulate cortices of the prefrontal cortex as well as discrete regions of the extended amygdala (i.e., basolateral) and extended basal ganglia (i.e., dorsolateral striatum, and caudal substantia nigra pars reticulata). These studies are the first to begin to elucidate circuitry by which this confirmed addiction-relevant QTL could influence behavior. This circuitry overlaps limbic circuitry involved in stress, providing additional mechanistic information. Alcdp1/Alcw1 maps to a region syntenic with human chromosome 1q, where multiple studies find significant associations with risk for alcoholism.
Collapse
Affiliation(s)
- Kari J Buck
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Gang Chen
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Laura B Kozell
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
40
|
Reilly MT, Noronha A, Goldman D, Koob GF. Genetic studies of alcohol dependence in the context of the addiction cycle. Neuropharmacology 2017; 122:3-21. [PMID: 28118990 DOI: 10.1016/j.neuropharm.2017.01.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 12/16/2022]
Abstract
Family, twin and adoption studies demonstrate clearly that alcohol dependence and alcohol use disorders are phenotypically complex and heritable. The heritability of alcohol use disorders is estimated at approximately 50-60% of the total phenotypic variability. Vulnerability to alcohol use disorders can be due to multiple genetic or environmental factors or their interaction which gives rise to extensive and daunting heterogeneity. This heterogeneity makes it a significant challenge in mapping and identifying the specific genes that influence alcohol use disorders. Genetic linkage and (candidate gene) association studies have been used now for decades to map and characterize genomic loci and genes that underlie the genetic vulnerability to alcohol use disorders. These approaches have been moderately successful in identifying several genes that contribute to the complexity of alcohol use disorders. Recently, genome-wide association studies have become one of the major tools for identifying genes for alcohol use disorders by examining correlations between millions of common single-nucleotide polymorphisms with diagnosis status. Genome-wide association studies are just beginning to uncover novel biology; however, the functional significance of results remains a matter of extensive debate and uncertainty. In this review, we present a select group of genome-wide association studies of alcohol dependence, as one example of a way to generate functional hypotheses, within the addiction cycle framework. This analysis may provide novel directions for validating the functional significance of alcohol dependence candidate genes. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Matthew T Reilly
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA.
| | - Antonio Noronha
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - David Goldman
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Chief, Laboratory of Neurogenetics, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - George F Koob
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Director NIAAA, 5635 Fishers Lane, Bethesda, MD 20852, USA
| |
Collapse
|
41
|
Seidemann T, Spies C, Morgenstern R, Wernecke KD, Netzhammer N. Influence of Volatile Anesthesia on the Release of Glutamate and other Amino Acids in the Nucleus Accumbens in a Rat Model of Alcohol Withdrawal: A Pilot Study. PLoS One 2017; 12:e0169017. [PMID: 28045949 PMCID: PMC5207639 DOI: 10.1371/journal.pone.0169017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/09/2016] [Indexed: 01/29/2023] Open
Abstract
Background Alcohol withdrawal syndrome is a potentially life-threatening condition, which can occur when patients with alcohol use disorders undergo general anesthesia. Excitatory amino acids, such as glutamate, act as neurotransmitters and are known to play a key role in alcohol withdrawal syndrome. To understand this process better, we investigated the influence of isoflurane, sevoflurane, and desflurane anesthesia on the profile of excitatory and inhibitory amino acids in the nucleus accumbens (NAcc) of alcohol-withdrawn rats (AWR). Methods Eighty Wistar rats were randomized into two groups of 40, pair-fed with alcoholic or non-alcoholic nutrition. Nutrition was withdrawn and microdialysis was performed to measure the activity of amino acids in the NAcc. The onset time of the withdrawal syndrome was first determined in an experiment with 20 rats. Sixty rats then received isoflurane, sevoflurane, or desflurane anesthesia for three hours during the withdrawal period, followed by one hour of elimination. Amino acid concentrations were measured using chromatography and results were compared to baseline levels measured prior to induction of anesthesia. Results Glutamate release increased in the alcohol group at five hours after the last alcohol intake (p = 0.002). After 140 min, desflurane anesthesia led to a lower release of glutamate (p < 0.001) and aspartate (p = 0.0007) in AWR compared to controls. GABA release under and after desflurane anesthesia was also significantly lower in AWR than controls (p = 0.023). Over the course of isoflurane anesthesia, arginine release decreased in AWR compared to controls (p < 0.001), and aspartate release increased after induction relative to controls (p20min = 0.015 and p40min = 0.006). However, amino acid levels did not differ between the groups as a result of sevoflurane anesthesia. Conclusions Each of three volatile anesthetics we studied showed different effects on excitatory and inhibitory amino acid concentrations. Under desflurane anesthesia, both glutamate and aspartate showed a tendency to be lower in AWR than controls over the whole timecourse. The inhibitory amino acid arginine increased in AWR compared to controls, whereas GABA levels decreased. However, there were no significant differences in amino acid concentrations under or after sevoflurane anesthesia. Under isoflurane, aspartate release increased in AWR following induction, and from 40 min to 140 min arginine release in controls was elevated. The precise mechanisms through which each of the volatile anesthetics affected amino acid concentrations are still unclear and further experimental research is required to draw reliable conclusions.
Collapse
Affiliation(s)
- Thomas Seidemann
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Spies
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| | - Rudolf Morgenstern
- Institute of Pharmacology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Nicolai Netzhammer
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
42
|
Kohno M, Dennis LE, McCready H, Hoffman WF. Executive Control and Striatal Resting-State Network Interact with Risk Factors to Influence Treatment Outcomes in Alcohol-Use Disorder. Front Psychiatry 2017; 8:182. [PMID: 28993741 PMCID: PMC5622290 DOI: 10.3389/fpsyt.2017.00182] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/11/2017] [Indexed: 12/20/2022] Open
Abstract
Alterations within mesocorticolimbic terminal regions commonly occur with alcohol use disorder (AUD). As pathological drug-seeking behavior may arise as a consequence of alcohol-induced neuroadaptations, it is critical to understand how such changes increase the likelihood of relapse. This report examined resting-state functional connectivity (RSFC) using both a seed-based and model-free approach in individuals in treatment for AUD and how dysregulation of network connectivity contributes to treatment outcomes. In order to provide a mechanism by which neural networks promote relapse, interactive effects of mesocorticolimbic connectivity and AUD risk factors in treatment completers and non-completers were examined. AUD group showed stronger RSFC between striatum, insula, and anterior cingulate cortex than controls. Within the AUD group, non-completers compared to completers showed enhanced RSFC between (1) striatum-insula, (2) executive control network (ECN)-amygdala, and (3) basal ganglia/salience network and striatum, precuneus, and insula. Completers showed enhanced RSFC between striatum-right dorsolateral prefrontal cortex. Furthermore, completers and non-completers differed in relationships between RSFC and relapse risk factors, where non-completers exhibited positive associations between craving intensity and RSFC of striatum-insula and ECN-amygdala. These findings provide evidence for interactions between corticolimbic connectivity in AUD and craving and establish an important link between network connectivity and dynamic risk factors that contribute to relapse. Results demonstrate that relapse vulnerability is attributed to craving dysregulation manifested by enhanced connectivity in striato-limbic regions and diminished corticostriatal connectivity.
Collapse
Affiliation(s)
- Milky Kohno
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Methamphetamine Abuse Research Center, Portland, OR, United States
| | - Laura E Dennis
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Methamphetamine Abuse Research Center, Portland, OR, United States
| | - Holly McCready
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Methamphetamine Abuse Research Center, Portland, OR, United States
| | - William F Hoffman
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States.,Methamphetamine Abuse Research Center, Portland, OR, United States.,Mental Health and Clinical Neurosciences Division, Portland, OR, United States.,Research Service Veterans Affairs Portland Healthcare System, Portland, OR, United States
| |
Collapse
|
43
|
Caruso JP, Susick LL, Charlton JL, Henson EL, Conti AC. Region-specific disruption of synapsin phosphorylation following ethanol administration in brain-injured mice. Brain Circ 2016; 2:183-188. [PMID: 30276296 PMCID: PMC6126228 DOI: 10.4103/2394-8108.195284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 11/11/2022] Open
Abstract
Introduction: Civilians and military personnel develop a range of physical and psychosocial impairments following traumatic brain injury (TBI), including alcohol abuse. As a consequence, increased rates of alcohol misuse magnify TBI-induced pathologies and impede rehabilitation efforts. Therefore, a developed understanding of the mechanisms that foster susceptibility of the injured brain to alcohol sensitivity and the response of the injured brain to alcohol is imperative for the treatment of TBI patients. Alcohol sensitivity has been demonstrated to be increased following experimental TBI and, in additional studies, regulated by presynaptic vesicle release mechanisms, including synapsin phosphorylation. Materials and Methods: Mice were exposed to controlled midline impact of the intact skull and assessed for cortical, hippocampal, and striatal expression of phosphorylated synapsin I and II in response to high-dose ethanol exposure administered 14 days following injury, a time point at which injured mice demonstrate increased sedation after ethanol exposure. Results and Discussion: Immunoblot quantitation revealed that TBI alone, compared to sham controls, significantly increased phosphorylated synapsin I and II protein expression in the striatum. In sham controls, ethanol administration significantly increased phosphorylated synapsin I and II protein expression compared to saline-treated sham controls; however, no significant increase in ethanol-induced phosphorylated synapsin I and II protein expression was observed in the striatum of injured mice compared to saline-treated TBI controls. A similar expression pattern was observed in the cortex although restricted to increases in phosphorylated synapsin II. Conclusion: These data show that increased phosphorylated synapsin expression in the injured striatum may reflect a compensatory neuroplastic response to TBI which is proposed to occur as a result of a compromised presynaptic response of the injured brain to high-dose ethanol. These results offer a mechanistic basis for the altered ethanol sensitivity observed following experimental TBI and contribute to our understanding of alcohol action in the injured brain.
Collapse
Affiliation(s)
- James P Caruso
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Laura L Susick
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Jennifer L Charlton
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Emily L Henson
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Alana C Conti
- John D. Dingell VA Medical Center and Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
44
|
Spoelder M, Hesseling P, Styles M, Baars AM, Lozeman-van 't Klooster JG, Lesscher HMB, Vanderschuren LJMJ. Dopaminergic neurotransmission in ventral and dorsal striatum differentially modulates alcohol reinforcement. Eur J Neurosci 2016; 45:147-158. [PMID: 27521051 DOI: 10.1111/ejn.13358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/14/2016] [Accepted: 08/02/2016] [Indexed: 01/18/2023]
Abstract
Dopaminergic neurotransmission in the striatum has been widely implicated in the reinforcing properties of substances of abuse. However, the striatum is functionally heterogeneous, and previous work has mostly focused on psychostimulant drugs. Therefore, we investigated how dopamine within striatal subregions modulates alcohol-directed behaviour in rats. We assessed the effects of infusion of the dopamine receptor antagonist alpha-flupenthixol into the shell and core of the nucleus accumbens (NAcc) and the dorsolateral striatum (DLS) on responding for alcohol under fixed ratio 1 (FR1) and progressive ratio (PR) schedules of reinforcement. Bilateral infusion of alpha-flupenthixol into the NAcc shell reduced responding for alcohol under both the FR1 (15 μg/side) and the PR schedule (3.75-15 μg/side) of reinforcement. Infusion of alpha-flupenthixol into the NAcc core (7.5-15 μg/side) also decreased responding for alcohol under both schedules. By contrast, alpha-flupenthixol infusion into the DLS did not affect FR1 responding, but reduced responding under the PR schedule (15 μg/side). The decreases in responding were related to earlier termination of responding during the session, whereas the onset and rate of responding remained largely unaffected. Together, these data suggest that dopamine in the NAcc shell is involved in the incentive motivation for alcohol, whereas DLS dopamine comes into play when obtaining alcohol requires high levels of effort. In contrast, NAcc core dopamine appears to play a more general role in alcohol reinforcement. In conclusion, dopaminergic neurotransmission acts in concert in subregions of the striatum to modulate different aspects of alcohol-directed behaviour.
Collapse
Affiliation(s)
- Marcia Spoelder
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Peter Hesseling
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Matthew Styles
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Annemarie M Baars
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - José G Lozeman-van 't Klooster
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Heidi M B Lesscher
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| |
Collapse
|
45
|
Yang X, Tian F, Zhang H, Zeng J, Chen T, Wang S, Jia Z, Gong Q. Cortical and subcortical gray matter shrinkage in alcohol-use disorders: a voxel-based meta-analysis. Neurosci Biobehav Rev 2016; 66:92-103. [PMID: 27108216 DOI: 10.1016/j.neubiorev.2016.03.034] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/17/2016] [Accepted: 03/24/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Xun Yang
- School of Sociality and Psychology, Southwest University for Nationalities, Chengdu 610041, China; Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Fangfang Tian
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Handi Zhang
- School of Sociality and Psychology, Southwest University for Nationalities, Chengdu 610041, China
| | - Jianguang Zeng
- School of Accounting, Southwestern University of Finance and Economics, Chengdu 611130, China
| | - Taolin Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Song Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhiyun Jia
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China; Department of Psychology, School of Public Administration, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Ayers-Ringler JR, Oliveros A, Qiu Y, Lindberg DM, Hinton DJ, Moore RM, Dasari S, Choi DS. Label-Free Proteomic Analysis of Protein Changes in the Striatum during Chronic Ethanol Use and Early Withdrawal. Front Behav Neurosci 2016; 10:46. [PMID: 27014007 PMCID: PMC4786553 DOI: 10.3389/fnbeh.2016.00046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/26/2016] [Indexed: 01/03/2023] Open
Abstract
The molecular mechanisms underlying the neuronal signaling changes in alcohol addiction and withdrawal are complex and multifaceted. The cortico-striatal circuit is highly implicated in these processes, and the striatum plays a significant role not only in the early stages of addiction, but in the developed-addictive state as well, including withdrawal symptoms. Transcriptional analysis is a useful method for determining changes in gene expression, however, the results do not always accurately correlate with protein levels. In this study, we employ label-free proteomic analysis to determine changes in protein expression within the striatum during chronic ethanol use and early withdrawal. The striatum, composed primarily of medium spiny GABAergic neurons, glutamatergic and dopaminergic nerve terminals and astrocytes, is relatively homogeneous for proteomic analysis. We were able to analyze more than 5000 proteins from both the dorsal (caudate and putamen) and ventral (nucleus accumbens) striatum and identified significant changes following chronic intermittent ethanol exposure and acute (8 h) withdrawal compared to ethanol naïve and ethanol exposure groups respectively. Our results showed significant changes in proteins involved in glutamate and opioid peptide signaling, and also uncovered novel pathways including mitochondrial function and lipid/cholesterol metabolism, as revealed by changes in electron transport chain proteins and RXR activation pathways. These results will be useful in the development of novel treatments for alcohol withdrawal and thereby aid in recovery from alcohol use disorder.
Collapse
Affiliation(s)
| | - Alfredo Oliveros
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Graduate School, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Yanyan Qiu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Graduate School, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Daniel M Lindberg
- Neurobiology of Disease PhD Program, Mayo Graduate School, Mayo Clinic Rochester, MN, USA
| | - David J Hinton
- Neurobiology of Disease PhD Program, Mayo Graduate School, Mayo Clinic Rochester, MN, USA
| | - Raymond M Moore
- Department of Biochemistry and Molecular Biology, Center for Individualized Medicine, Mayo Clinic Rochester, MN, USA
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic College of Medicine Rochester, MN, USA
| | - Doo-Sup Choi
- Neurobiology of Disease PhD Program, Mayo Graduate School, Mayo ClinicRochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Graduate School, Mayo Clinic College of MedicineRochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic College of MedicineRochester, MN, USA
| |
Collapse
|
47
|
McKim TH, Shnitko TA, Robinson DL, Boettiger CA. Translational Research on Habit and Alcohol. CURRENT ADDICTION REPORTS 2016; 3:37-49. [PMID: 26925365 DOI: 10.1007/s40429-016-0089-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Habitual actions enable efficient daily living, but they can also contribute to pathological behaviors that resistant change, such as alcoholism. Habitual behaviors are learned actions that appear goal-directed but are in fact no longer under the control of the action's outcome. Instead, these actions are triggered by stimuli, which may be exogenous or interoceptive, discrete or contextual. A major hallmark characteristic of alcoholism is continued alcohol use despite serious negative consequences. In essence, although the outcome of alcohol seeking and drinking is dramatically devalued, these actions persist, often triggered by environmental cues associated with alcohol use. Thus, alcoholism meets the definition of an initially goal-directed behavior that converts to a habit-based process. Habit and alcohol have been well investigated in rodent models, with comparatively less research in non-human primates and people. This review focuses on translational research on habit and alcohol with an emphasis on cross-species methodology and neural circuitry.
Collapse
Affiliation(s)
- Theresa H McKim
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Davie Hall, CB #3270, Chapel Hill, NC 27599
| | - Tatiana A Shnitko
- University of North Carolina at Chapel Hill, Bowles Center for Alcohol Studies, CB #7178, Chapel Hill, NC 27599
| | - Donita L Robinson
- University of North Carolina at Chapel Hill, Department of Psychiatry, Bowles Center for Alcohol Studies, CB #7178, Chapel Hill, NC 27599
| | - Charlotte A Boettiger
- Biomedical Research Imaging Center, Bowles Center for Alcohol Studies, Davie Hall, CB #3270, Chapel Hill, NC 27599
| |
Collapse
|
48
|
Guan YZ, Ye JH. Glycine blocks long-term potentiation of GABAergic synapses in the ventral tegmental area. Neuroscience 2016; 318:134-42. [PMID: 26806277 DOI: 10.1016/j.neuroscience.2016.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 10/22/2022]
Abstract
The mesocorticolimbic dopamine system, originating in the ventral tegmental area (VTA) is normally constrained by GABA-mediated synaptic inhibition. Accumulating evidence indicates that long-term potentiation of GABAergic synapses (LTPGABA) in VTA dopamine neurons plays an important role in the actions of drugs of abuse, including ethanol. We previously showed that a single infusion of glycine into the VTA of rats strongly reduces ethanol intake for 24h. In the current study, we examined the effect of glycine on the electrophysiological activities of putative dopamine VTA neurons in midbrain slices from ethanol-naïve rats. We report here that a 15-min exposure to 10 μM glycine prevented trains of high-frequency stimulation (HFS) from producing LTPGABA, which was rescued by the glycine receptor (GlyR) antagonist strychnine. Glycine also concentration-dependently decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs). By contrast, glycine pretreatment did not prevent potentiation of inhibitory postsynaptic currents (IPSCs) during a continuous exposure to the nitric oxide (NO) donor, SNAP (S-nitroso-N-acetylpenicillamine), or a brief exposure to 10 μM glycine and 10 μM NMDA (N-methyl-D-aspartate), an agonist of NMDA-type glutamate receptors. Thus, the blockade of LTPGABA by glycine is probably resulted from suppressing glutamate release by activating the GlyRs on the glutamatergic terminals. This effect of glycine may contribute to the reduction in ethanol intake induced by intra-VTA glycine observed in vivo.
Collapse
Affiliation(s)
- Y-Z Guan
- Department of Anesthesiology, Pharmacology and Physiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA; Department of Physiology, Mudanjiang Medical University, Mudanjiang, China.
| | - J-H Ye
- Department of Anesthesiology, Pharmacology and Physiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
49
|
Clarke RBC, Söderpalm B, Lotfi A, Ericson M, Adermark L. Involvement of Inhibitory Receptors in Modulating Dopamine Signaling and Synaptic Activity Following Acute Ethanol Exposure in Striatal Subregions. Alcohol Clin Exp Res 2015; 39:2364-74. [DOI: 10.1111/acer.12895] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/01/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Rhona B. C. Clarke
- Addiction Biology Unit; Institute of Neuroscience and Physiology; Department of Psychiatry and Neurochemistry; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | - Bo Söderpalm
- Addiction Biology Unit; Institute of Neuroscience and Physiology; Department of Psychiatry and Neurochemistry; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
- Beroendekliniken; Sahlgrenska University Hospital; Gothenburg Sweden
| | - Amir Lotfi
- Addiction Biology Unit; Institute of Neuroscience and Physiology; Department of Psychiatry and Neurochemistry; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | - Mia Ericson
- Addiction Biology Unit; Institute of Neuroscience and Physiology; Department of Psychiatry and Neurochemistry; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | - Louise Adermark
- Addiction Biology Unit; Institute of Neuroscience and Physiology; Department of Psychiatry and Neurochemistry; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
50
|
Kim JI, Ganesan S, Luo SX, Wu YW, Park E, Huang EJ, Chen L, Ding JB. Aldehyde dehydrogenase 1a1 mediates a GABA synthesis pathway in midbrain dopaminergic neurons. Science 2015; 350:102-6. [PMID: 26430123 DOI: 10.1126/science.aac4690] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Midbrain dopamine neurons are an essential component of the basal ganglia circuitry, playing key roles in the control of fine movement and reward. Recently, it has been demonstrated that γ-aminobutyric acid (GABA), the chief inhibitory neurotransmitter, is co-released by dopamine neurons. Here, we show that GABA co-release in dopamine neurons does not use the conventional GABA-synthesizing enzymes, glutamate decarboxylases GAD65 and GAD67. Our experiments reveal an evolutionarily conserved GABA synthesis pathway mediated by aldehyde dehydrogenase 1a1 (ALDH1a1). Moreover, GABA co-release is modulated by ethanol (EtOH) at concentrations seen in blood alcohol after binge drinking, and diminished ALDH1a1 leads to enhanced alcohol consumption and preference. These findings provide insights into the functional role of GABA co-release in midbrain dopamine neurons, which may be essential for reward-based behavior and addiction.
Collapse
Affiliation(s)
- Jae-Ick Kim
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Subhashree Ganesan
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sarah X Luo
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA. Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yu-Wei Wu
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Esther Park
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA. Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA. Pathology Service 113B, San Francisco VA Medical Center, San Francisco, CA 94121, USA
| | - Lu Chen
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA. Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|