1
|
de Bejczy A, Addolorato G, Aubin HJ, Guiraud J, Korpi ER, John Nutt D, Witkiewitz K, Söderpalm B. AUD in perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:1-19. [PMID: 38555113 DOI: 10.1016/bs.irn.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Alcohol is a major cause of pre-mature death and individual suffering worldwide, and the importance of diagnosing and treating AUD cannot be overstated. Given the global burden and the high attributable factor of alcohol in a vast number of diseases, the need for additional interventions and the development of new medicines is considered a priority by the World Health Organization (WHO). As of today, AUD is severely under-treated with a treatment gap nearing 90%, strikingly higher than that for other psychiatric disorders. Patients often seek treatment late in the progress of the disease and even among those who seek treatment only a minority receive medication, mirroring the still-prevailing stigma of the disease, and a lack of access to effective treatments, as well as a reluctance to total abstinence. To increase adherence, treatment goals should focus not only on maintaining abstinence, but also on harm reduction and psychosocial functioning. A personalised approach to AUD treatment, with a holistic view, and tailored therapy has the potential to improve AUD treatment outcomes by targeting the heterogeneity in genetics and pathophysiology, as well as reason for, and reaction to drinking. Also, the psychiatric co-morbidity rates are high in AUD and dual diagnosis can worsen symptoms and influence treatment response and should be considered in the treatment strategies.
Collapse
Affiliation(s)
- Andrea de Bejczy
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Giovanni Addolorato
- Department of Medical and Surgical Sciences, Università Cattolica di Roma, Rome, Italy; Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Henri-Jean Aubin
- Université Paris-Saclay, Inserm, CESP, Villejuif, France; AP-HP, Université Paris Saclay, Villejuif, France
| | - Julien Guiraud
- Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands; Vergio, Clichy, France
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David John Nutt
- Imperial College London and GABA Labs, London, United Kingdom
| | - Katie Witkiewitz
- Department of Psychology and Center on Alcohol, Substance Use, and Addictions, University of New Mexico, Albuquerque, New Mexico, United States
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
2
|
Farahbakhsh ZZ, Holleran KM, Sens JP, Fordahl SC, Mauterer MI, López AJ, Cuzon Carlson VC, Kiraly DD, Grant KA, Jones SR, Siciliano CA. Synchrony between midbrain gene transcription and dopamine terminal regulation is modulated by chronic alcohol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.584711. [PMID: 38559169 PMCID: PMC10979957 DOI: 10.1101/2024.03.15.584711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alcohol use disorder is marked by disrupted behavioral and emotional states which persist into abstinence. The enduring synaptic alterations that remain despite the absence of alcohol are of interest for interventions to prevent relapse. Here, 28 male rhesus macaques underwent over 20 months of alcohol drinking interspersed with three 30-day forced abstinence periods. After the last abstinence period, we paired direct sub-second dopamine monitoring via ex vivo voltammetry in nucleus accumbens slices with RNA-sequencing of the ventral tegmental area. We found persistent augmentation of dopamine transporter function, kappa opioid receptor sensitivity, and dynorphin release - all inhibitory regulators which act to decrease extracellular dopamine. Surprisingly, though transcript expression was not altered, the relationship between gene expression and functional readouts of these encoded proteins was highly dynamic and altered by drinking history. These results outline the long-lasting synaptic impact of alcohol use and suggest that assessment of transcript-function relationships is critical for the rational design of precision therapeutics.
Collapse
|
3
|
Zheng L, Aimaiti Z, Long L, Xia C, Wang W, Zhou ZZ. Discovery of 4-Ethoxy-6-chloro-5-azaindazoles as Novel PDE4 Inhibitors for the Treatment of Alcohol Use Disorder and Alcoholic Liver Diseases. J Med Chem 2024; 67:728-753. [PMID: 38156615 DOI: 10.1021/acs.jmedchem.3c02087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Alcohol use disorder (AUD) results in numerous disabilities and approximately 3 million deaths annually, caused mainly by alcoholic liver disease (ALD). Phosphodiesterase IV (PDE4) has emerged as an attractive molecular target for a new treatment for AUD and ALD. In this study, we describe the identification of 5-azaindazole analogues as PDE4 inhibitors against AUD and ALD. System optimization studies led to the discovery of ZL40 (IC50 = 37.4 nM) with a remarkable oral bioavailability (F = 94%), satisfactory safety, and a lower emetogenic potency than the approved PDE4 inhibitors roflumilast and apremilast. Encouragingly, ZL40 exhibited AUD therapeutic effects by decreasing alcohol intake and improving acute alcohol-induced sedation and motor impairment. Meanwhile, ZL40 displayed the potential to alleviate alcoholic liver injury and attenuate inflammation in the NIAAA mice model. These results showed that ZL40 is a promising compound for future drug development to treat alcohol-related diseases.
Collapse
Affiliation(s)
- Lei Zheng
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zulihuma Aimaiti
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lu Long
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chuang Xia
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenya Wang
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhong-Zhen Zhou
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
4
|
McArdle CJ, Arnone AA, Heaney CF, Raab-Graham KF. A paradoxical switch: the implications of excitatory GABAergic signaling in neurological disorders. Front Psychiatry 2024; 14:1296527. [PMID: 38268565 PMCID: PMC10805837 DOI: 10.3389/fpsyt.2023.1296527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. In the mature brain, inhibitory GABAergic signaling is critical in maintaining neuronal homeostasis and vital human behaviors such as cognition, emotion, and motivation. While classically known to inhibit neuronal function under physiological conditions, previous research indicates a paradoxical switch from inhibitory to excitatory GABAergic signaling that is implicated in several neurological disorders. Various mechanisms have been proposed to contribute to the excitatory switch such as chloride ion dyshomeostasis, alterations in inhibitory receptor expression, and modifications in GABAergic synaptic plasticity. Of note, the hypothesized mechanisms underlying excitatory GABAergic signaling are highlighted in a number of neurodevelopmental, substance use, stress, and neurodegenerative disorders. Herein, we present an updated review discussing the presence of excitatory GABAergic signaling in various neurological disorders, and their potential contributions towards disease pathology.
Collapse
Affiliation(s)
- Colin J. McArdle
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Alana A. Arnone
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Chelcie F. Heaney
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Khikhmetova K, Semenova Y, Bjørklund G. The Roles of Endogenous D2R Dopamine and μ-opioid Receptors of the Brain in Alcohol use Disorder. Curr Med Chem 2024; 31:6393-6406. [PMID: 37921171 DOI: 10.2174/0109298673248999231013043249] [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: 02/13/2023] [Revised: 09/02/2023] [Accepted: 09/15/2023] [Indexed: 11/04/2023]
Abstract
Alcohol use disorder (AUD) affects millions of people worldwide. It is characterized by a strong physiological and psychological craving to consume large amounts of alcohol despite adverse consequences. Alcohol use disorder carries a large health and economic burden on society. Despite its prevalence, AUD is still severely undertreated. The precise molecular mechanisms of how alcohol addiction forms are yet unknown. However, previous studies on animal models show that along with the μ-opioid receptors, the D2R dopamine receptors may also be involved in alcohol craving and reward pathways. Currently, there is a limited number of treatment strategies for alcohol use disorder, which include several medications and therapy. By understanding the limitations of current treatment options and exploring new potential targets, it could be possible to find more effective ways of treating AUD in the future.
Collapse
Affiliation(s)
| | - Yuliya Semenova
- School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
6
|
Bremmer MP, Hendershot CS. Social Media as Pharmacovigilance: The Potential for Patient Reports to Inform Clinical Research on Glucagon-Like Peptide 1 (GLP-1) Receptor Agonists for Substance Use Disorders. J Stud Alcohol Drugs 2024; 85:5-11. [PMID: 37917019 PMCID: PMC10846600 DOI: 10.15288/jsad.23-00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/03/2023] Open
Abstract
The surge in popularity of semaglutide (Ozempic, Wegovy, Rybelsus) and other glucagon-like-peptide 1 (GLP-1) receptor agonists has been accompanied by widespread reports of unintended reductions in alcohol use (and other addictive behaviors) during treatment. With clinical trials of GLP-1 receptor agonists for substance use only recently under way, anecdotal reports (including via social media) are now a primary reason for interest in potential effects of GLP-1 receptor agonists on alcohol use in patient populations. The nature and volume of these reports raises the prospect that social media data can potentially be leveraged to inform the study of novel addiction treatments and the prioritization of behavioral or neurobiological targets for mechanistic research. This approach, which aligns with recent efforts to apply social media data to pharmacovigilance, may be particularly relevant for drug repurposing efforts. This possibility is illustrated by a thematic analysis of anonymous online reports concerning changes in alcohol use or alcohol-related effects during treatment with GLP-1 receptor agonists. These reports not only support the rationale for clinical trials but also point to potential neurobehavioral mechanisms (e.g., satiety, craving/preoccupation, aversion, altered subjective response) that might inform hypotheses for human laboratory and neuroscience studies. Refined methods for capturing patient reports of incidental medication effects on addictive behaviors at large scale could potentially lead to novel, pharmacovigilance-based approaches to identify candidate therapies for drug repurposing efforts.
Collapse
Affiliation(s)
- Michael P. Bremmer
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian S. Hendershot
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
7
|
Jiang Z, Chen Z, Chen X. Candidate gene-environment interactions in substance abuse: A systematic review. PLoS One 2023; 18:e0287446. [PMID: 37906564 PMCID: PMC10617739 DOI: 10.1371/journal.pone.0287446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 06/06/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND The abuse of psychogenic drugs can lead to multiple health-related problems. Genetic and environmental vulnerabilities are factors in the emergence of substance use disorders. Empirical evidence regarding the gene-environment interaction in substance use is mixed. Summaries of the latest findings from a candidate gene approach will be useful for revealing the significance of particular gene contributions. Thus, we aim to identify different gene-environment interactions in patterns of substance use and investigate whether any effects trend notably across different genders and races. METHODS We reviewed published studies, until March 1, 2022, on substance use for candidate gene-environment interaction. Basic demographics of the included studies, target genes, environmental factors, main findings, patterns of gene-environment interaction, and other relevant information were collected and summarized. RESULTS Among a total of 44 studies, 38 demonstrated at least one significant interaction effect. About 61.5% of studies on the 5-HTTLPR gene, 100% on the MAOA gene, 42.9% on the DRD2 gene, 50% on the DRD4 gene, 50% on the DAT gene, 80% on the CRHR1 gene, 100% on the OPRM1 gene, 100% on the GABRA1 gene, and 50% on the CHRNA gene had a significant gene-environment interaction effect. The diathesis-stress model represents a dominant interaction pattern (89.5%) in the studies with a significant interaction effect; the remaining significant effect on substance use is found in the differential susceptibility model. The social push and swing model were not reported in the included studies. CONCLUSION The gene-environment interaction research on substance use behavior is methodologically multidimensional, which causes difficulty in conducting pooled analysis, or stated differently-making it hard to identify single sources of significant influence over maladaptive patterns of drug taking. In decreasing the heterogeneity and facilitating future pooled analysis, researchers must (1) replicate the existing studies with consistent study designs and measures, (2) conduct power calculations to report gene-environment correlations, (3) control for covariates, and (4) generate theory-based hypotheses with factorial based experiments when designing future studies.
Collapse
Affiliation(s)
- Zheng Jiang
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zidong Chen
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xi Chen
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Department of Sociology and Social Policy, Lingnan University, Tuen Mun, Hong Kong
| |
Collapse
|
8
|
Gowin JL, Sloan ME, Kirk-Provencher KT, Rosenblatt SL, Penner AE, Stangl BL, Byrd ND, Swan JE, Ramchandani VA. Opioid receptor antagonism and neural response to monetary rewards: Pilot studies in light and heavy alcohol users. J Psychopharmacol 2023; 37:937-941. [PMID: 37530456 DOI: 10.1177/02698811231191707] [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] [Indexed: 08/03/2023]
Abstract
Alcohol use disorder (AUD) is a prevalent condition associated with high degree of comorbidity and mortality. Among the few approved pharmacotherapies for AUD, two involve opioid receptor antagonism. Naltrexone and nalmefene are thought to act via opioid receptor blockage to reduce neural response to alcohol and drug-associated cues and consumption, but there have been limited efforts to characterize these effects in humans. In these studies, we sought to test the magnitude of opioid antagonism effects on neural response to monetary rewards in two groups: light drinkers (for the naltrexone study) and heavy drinkers (for the nalmefene study). We conducted double-blind, randomized, crossover pilot studies of reward activation in the brain following acute administration of opioid antagonist and placebo in 11 light and 9 heavy alcohol users. We used a monetary incentive delay task during functional MRI. We found a main effect of cue type on BOLD activation in the nucleus accumbens, demonstrating a neural reward response. The effect of opioid antagonism, relative to placebo, was small and nonsignificant for reward activation in the accumbens for both light and heavy alcohol users. Based on the results of two pilot studies, opioid antagonist medications do not appear to decrease neural activation to monetary rewards in the nucleus accumbens relative to placebo.
Collapse
Affiliation(s)
- Joshua L Gowin
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew E Sloan
- Addictions Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | | | - Sophie L Rosenblatt
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anne E Penner
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bethany L Stangl
- National Institute on Alcohol Abuse and Alcoholism, Laboratory on Human Psychopharmacology, Bethesda, MD, USA
| | - Nia D Byrd
- National Institute on Alcohol Abuse and Alcoholism, Laboratory on Human Psychopharmacology, Bethesda, MD, USA
| | - Julia E Swan
- National Institute on Alcohol Abuse and Alcoholism, Laboratory on Human Psychopharmacology, Bethesda, MD, USA
| | - Vijay A Ramchandani
- National Institute on Alcohol Abuse and Alcoholism, Laboratory on Human Psychopharmacology, Bethesda, MD, USA
| |
Collapse
|
9
|
Alshehri FS. A Review of the Characteristics of Clinical Trials and Potential Medications for Alcohol Dependence: Data Analysis from ClinicalTrials.gov. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1101. [PMID: 37374305 DOI: 10.3390/medicina59061101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/20/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
Objective. This study provides a comprehensive analysis of the characteristics of clinical trials related to alcohol dependence that are registered on ClinicalTrials.gov. Methods. All ClinicalTrials.gov trials registered up to 1 January 2023 were examined, focusing on trials that involved alcohol dependence. All 1295 trials were summarized by presenting their characteristics and results and reviewed most intervention drugs used in the treatment of alcohol dependence. Results. The study analysis identified a total of 1295 clinical trials registered on ClinicalTrials.gov that were focused on alcohol dependence. Of these, 766 trials had been completed, representing 59.15% of the total, while 230 trials were currently recruiting participants, accounting for 17.76% of the total. None of the trials had yet been approved for marketing. The majority of the studies included in this analysis were interventional studies (1145 trials, or 88.41%), which accounted for most of the patients enrolled in the trials. In contrast, observational studies represented only a small portion of the trials (150 studies, or 11.58%) and involved a smaller number of patients. In terms of geographic distribution, the majority of registered studies were located in North America (876 studies, or 67.64%), while only a small number of studies were registered in South America (7 studies, or 0.54%). Conclusions. The purpose of this review is to provide a basis for the treatment of alcohol dependence and prevention of its onset through an overview of clinical trials registered at ClinicalTrials.gov. It also offers essential information for future research to guide future studies.
Collapse
Affiliation(s)
- Fahad S Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| |
Collapse
|
10
|
Rahman MT, Decker AM, Ben Hamida S, Perrey DA, Chaminda Lakmal HH, Maitra R, Darcq E, Kieffer BL, Jin C. Improvement of the Metabolic Stability of GPR88 Agonist RTI-13951-33: Design, Synthesis, and Biological Evaluation. J Med Chem 2023; 66:2964-2978. [PMID: 36749855 PMCID: PMC9974843 DOI: 10.1021/acs.jmedchem.2c01983] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
GPR88 is an orphan G protein-coupled receptor mainly expressed in the brain, whose endogenous ligand has not yet been identified. To elucidate GPR88 functions, our group has developed RTI-13951-33 (1b) as the first in vivo active GPR88 agonist, but its poor metabolic stability and moderate brain permeability remain to be further optimized. Here, we report the design, synthesis, and pharmacological characterization of a new series of RTI-13951-33 analogues with the aim of improving pharmacokinetic properties. As a result, we identified a highly potent GPR88 agonist RTI-122 (30a) (cAMP EC50 = 11 nM) with good metabolic stability (half-life of 5.8 h) and brain permeability (brain/plasma ratio of >1) in mice. Notably, RTI-122 was more effective than RTI-13951-33 in attenuating the binge-like alcohol drinking behavior in the drinking-in-the-dark paradigm. Collectively, our findings suggest that RTI-122 is a promising lead compound for drug discovery research of GPR88 agonists.
Collapse
Affiliation(s)
- Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Sami Ben Hamida
- INSERM UMR 1247, University of Picardie Jules Verne, Amiens 80025, France
| | - David A Perrey
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Hetti Handi Chaminda Lakmal
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Emmanuel Darcq
- INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | | | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
11
|
MacKillop J, Agabio R, Feldstein Ewing SW, Heilig M, Kelly JF, Leggio L, Lingford-Hughes A, Palmer AA, Parry CD, Ray L, Rehm J. Hazardous drinking and alcohol use disorders. Nat Rev Dis Primers 2022; 8:80. [PMID: 36550121 PMCID: PMC10284465 DOI: 10.1038/s41572-022-00406-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2022] [Indexed: 12/24/2022]
Abstract
Alcohol is one of the most widely consumed psychoactive drugs globally. Hazardous drinking, defined by quantity and frequency of consumption, is associated with acute and chronic morbidity. Alcohol use disorders (AUDs) are psychiatric syndromes characterized by impaired control over drinking and other symptoms. Contemporary aetiological perspectives on AUDs apply a biopsychosocial framework that emphasizes the interplay of genetics, neurobiology, psychology, and an individual's social and societal context. There is strong evidence that AUDs are genetically influenced, but with a complex polygenic architecture. Likewise, there is robust evidence for environmental influences, such as adverse childhood exposures and maladaptive developmental trajectories. Well-established biological and psychological determinants of AUDs include neuroadaptive changes following persistent use, differences in brain structure and function, and motivational determinants including overvaluation of alcohol reinforcement, acute effects of environmental triggers and stress, elevations in multiple facets of impulsivity, and lack of alternative reinforcers. Social factors include bidirectional roles of social networks and sociocultural influences, such as public health control strategies and social determinants of health. An array of evidence-based approaches for reducing alcohol harms are available, including screening, pharmacotherapies, psychological interventions and policy strategies, but are substantially underused. Priorities for the field include translating advances in basic biobehavioural research into novel clinical applications and, in turn, promoting widespread implementation of evidence-based clinical approaches in practice and health-care systems.
Collapse
Affiliation(s)
- James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada.
- Homewood Research Institute, Guelph, ON, Canada.
| | - Roberta Agabio
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- Neuroscience Institute, Section of Cagliari, National Research Council, Cagliari, Italy
| | - Sarah W Feldstein Ewing
- Department of Psychology, University of Rhode Island, Kingston, RI, USA
- Department of Psychology and Behavioural Sciences, Centre for Alcohol and Drug Research, Aarhus University, Aarhus, Denmark
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - John F Kelly
- Recovery Research Institute and Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Anne Lingford-Hughes
- Division of Psychiatry, Imperial College London, London, UK
- Central North West London NHS Foundation Trust, London, UK
| | - Abraham A Palmer
- Department of Psychiatry & Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Charles D Parry
- Alcohol, Tobacco and Other Drug Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - Lara Ray
- Departments of Psychology and Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jürgen Rehm
- Institute for Mental Health Policy Research, Campbell Family Mental Health Research Institute, PAHO/WHO Collaborating Centre, Centre for Addiction and Mental Health, Toronto, Canada
- Dalla Lana School of Public Health; Institute of Health Policy, Management and Evaluation; & Department of Psychiatry, University of Toronto (UofT), Toronto, Canada
- WHO European Region Collaborating Centre at Public Health Institute of Catalonia, Barcelona, Spain
- Technische Universität Dresden, Klinische Psychologie & Psychotherapie, Dresden, Germany
- Department of International Health Projects, Institute for Leadership and Health Management, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Zentrum für Interdisziplinäre Suchtforschung der Universität Hamburg (ZIS), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Jiang W, Chen J, Vidjro OE, Zhang Y, Guo G, Li Z, Qi Y, Dai R, Ma T. Construction and evaluation of an alcohol vapor chamber system. J Biomed Res 2022; 37:115-124. [PMID: 36529969 PMCID: PMC10018410 DOI: 10.7555/jbr.36.20220151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An increasing number of studies demonstrated that alcohol vapor chamber is an effective way to model physical signs of alcohol use disorders. Although researchers are developing different vapor chambers to study chronic alcohol exposure model worldwide, few studies build and modify their own vapor chambers in China. Here, we designed and established an alcohol vapor chamber system for small animals. We described a paradigm showing how to control and monitor alcohol concentration in whole system. The vapor chamber system with several advantages including accommodating up to ten standard mouse cages. Furthermore, the system was tested by evaluating the blood alcohol concentration and neuron injury in mice. Importantly, the alcohol withdrawal after vapor exposure caused motor coordination impairment, anxiolytic- and depression-like behavior. Finally, the N-methyl-D-aspartate receptor (NMDAR)-mediated glutamatergic transmissions in the medial prefrontal cortex was changed after alcohol vapor exposure-induced behaviors. The frequency and amplitude of spontaneous excitatory postsynaptic currents between control and alcohol groups were not different, suggesting that alcohol exposure-induced behaviors are associated with the change in NMDAR response. Taken together, the new alcohol vapor chamber system was constructed, which would help to research the relationship between the stable alcohol exposure and withdrawal behaviors and to study chronic alcohol exposure-induced disorders in China.
Collapse
Affiliation(s)
- Wan Jiang
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiajia Chen
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Olivia Ewi Vidjro
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Yingying Zhang
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Gengni Guo
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ziyi Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Yize Qi
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Rouli Dai
- National Institute of Drug Clinical Trial, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210031, China
| | - Tengfei Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| |
Collapse
|
13
|
Antonelli M, Sestito L, Tarli C, Addolorato G. Perspectives on the pharmacological management of alcohol use disorder: Are the approved medications effective? Eur J Intern Med 2022; 103:13-22. [PMID: 35597734 DOI: 10.1016/j.ejim.2022.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/18/2022]
Abstract
INTRODUCTION In the last decades, many medications have been tested for the treatment of Alcohol Use Disorder (AUD). Among them, disulfiram, acamprosate, naltrexone, nalmefene, sodium oxybate and baclofen have been approved in different countries, with different specific indications. Topiramate is not approved for the treatment of AUD, however, it is suggested as a therapeutic option by the American Psychiatric Association for patients who do not tolerate or respond to approved therapies. AREAS COVERED In this narrative review we have analyzed the main studies available in literature, investigating the efficacy and safety of these medications, distinguishing whether they were oriented towards abstinence or not. Randomized controlled studies, analyzing larger populations for longer periods were the main focus of our analysis. CONCLUSIONS The medications currently available for the treatment of AUD are quite effective, yet further progress can still be achieved through the personalized strategies. Also, these medications are still markedly underutilized in clinical practice and many patients do not have access to specialized treatment.
Collapse
Affiliation(s)
- Mariangela Antonelli
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luisa Sestito
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudia Tarli
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Addolorato
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy.
| |
Collapse
|
14
|
Substance use, microbiome and psychiatric disorders. Pharmacol Biochem Behav 2022; 219:173432. [PMID: 35905802 DOI: 10.1016/j.pbb.2022.173432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/29/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
Abstract
Accumulating evidence from several studies has shown association between substance use, dysregulation of the microbiome and psychiatric disorders such as depression, anxiety, and psychosis. Many of the abused substances such as cocaine and alcohol have been shown to alter immune signaling pathways and cause inflammation in both the periphery and the central nervous system (CNS). In addition, these substances of abuse also alter the composition and function of the gut microbiome which is known to play important roles such as the synthesis of neurotransmitters and metabolites, that affect the CNS homeostasis and consequent behavioral outcomes. The emerging interactions between substance use, microbiome and CNS neurochemical alterations could contribute to the development of psychiatric disorders. This review provides an overview of the associative effects of substance use such as alcohol, cocaine, methamphetamine, nicotine and opioids on the gut microbiome and psychiatric disorders involving anxiety, depression and psychosis. Understanding the relationship between substance use, microbiome and psychiatric disorders will provide insights for potential therapeutic targets, aimed at mitigating these adverse outcomes.
Collapse
|
15
|
Choi MR, Cho S, Kim DJ, Choi JS, Jin YB, Kim M, Chang HJ, Jeon SH, Yang YD, Lee SR. Effects of Ethanol on Expression of Coding and Noncoding RNAs in Murine Neuroblastoma Neuro2a Cells. Int J Mol Sci 2022; 23:ijms23137294. [PMID: 35806296 PMCID: PMC9267046 DOI: 10.3390/ijms23137294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Excessive use of alcohol can induce neurobiological and neuropathological alterations in the brain, including the hippocampus and forebrain, through changes in neurotransmitter systems, hormonal systems, and neuroimmune processes. We aimed to investigate the effects of ethanol on the expression of coding and noncoding RNAs in a brain-derived cell line exposed to ethanol. After exposing Neuro2a cells, a neuroblastoma cell line, to ethanol for 24 and 72 h, we observed cell proliferation and analyzed up- and downregulated mRNAs and long noncoding RNAs (lncRNAs) using total RNA-Seq technology. We validated the differential expression of some mRNAs and lncRNAs by RT-qPCR and analyzed the expression of Cebpd and Rnu3a through knock-down of Cebpd. Cell proliferation was significantly reduced in cells exposed to 100 mM ethanol for 72 h, with 1773 transcripts up- or downregulated by greater than three-fold in ethanol-treated cells compared to controls. Of these, 514 were identified as lncRNAs. Differentially expressed mRNAs and lncRNAs were mainly observed in cells exposed to ethanol for 72 h, in which Atm and Cnr1 decreased, but Trib3, Cebpd, and Spdef increased. On the other hand, lncRNAs Kcnq1ot1, Tug1, and Xist were changed by ethanol, and Rnu3a in particular was greatly increased by chronic ethanol treatment through inhibition of Cebpd. Our results increase the understanding of cellular and molecular mechanisms related to coding and noncoding RNAs in an in vitro model of acute and chronic exposure to ethanol.
Collapse
Affiliation(s)
- Mi Ran Choi
- Laboratory Animal Research Center, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Sinyoung Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jung-Seok Choi
- Department of Psychiatry, Samsung Medical Center, Seoul 06351, Korea;
| | - Yeung-Bae Jin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Miran Kim
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon 16499, Korea; (M.K.); (H.J.C.)
| | - Hye Jin Chang
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon 16499, Korea; (M.K.); (H.J.C.)
| | - Seong Ho Jeon
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
| | - Young Duk Yang
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
- Correspondence: (Y.D.Y.); (S.-R.L.); Tel.: +82-31-881-7170 (Y.D.Y.); +82-31-219-4499 (S.-R.L.)
| | - Sang-Rae Lee
- Laboratory Animal Research Center, Ajou University School of Medicine, Suwon 16499, Korea;
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Correspondence: (Y.D.Y.); (S.-R.L.); Tel.: +82-31-881-7170 (Y.D.Y.); +82-31-219-4499 (S.-R.L.)
| |
Collapse
|
16
|
Tarli C, Mirijello A, Addolorato G. Treating Alcohol Use Disorder in Patients with Alcohol-Associated Liver Disease: Controversies in Pharmacological Therapy. Semin Liver Dis 2022; 42:138-150. [PMID: 35292951 DOI: 10.1055/a-1798-2872] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alcohol use disorder (AUD) is one of the main causes of global death and disability. The liver represents the main target of alcohol damage, and alcohol-associated liver disease (ALD) represents the first cause of liver cirrhosis in Western countries. Alcohol abstinence is the main goal of treatment in AUD patients with ALD, as treatments for ALD are less effective when drinking continues. Moreover, the persistence of alcohol consumption is associated with higher mortality, increased need for liver transplantation, and graft loss. The most effective treatment for AUD is the combination of psychosocial interventions, pharmacological therapy, and medical management. However, the effectiveness of these treatments in patients with ALD is doubtful even because AUD patients with ALD are usually excluded from pharmacological trials due to concerns on liver safety. This narrative review will discuss the treatment options for AUD-ALD patients focusing on controversies in pharmacological therapy.
Collapse
Affiliation(s)
- Claudia Tarli
- Internal Medicine and Alcohol Related Disease Unit, Department of Medical and Surgical Sciences, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, Rome, Italy
| | - Antonio Mirijello
- Internal Medicine Unit, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giovanni Addolorato
- Internal Medicine and Alcohol Related Disease Unit, Department of Medical and Surgical Sciences, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Rome, Rome, Italy.,CEMAD Digestive Disease Center, Department of Medical and Surgical Sciences, Hepatology and Gastroenterology Unit, Catholic University of Rome, l.go Gemelli, Rome, Italy.,Internal Medicine Unit, Department of Internal Medicine and Gastroenterology, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A.Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Graham DP, Harding MJ, Nielsen DA. Pharmacogenetics of Addiction Therapy. Methods Mol Biol 2022; 2547:437-490. [PMID: 36068473 DOI: 10.1007/978-1-0716-2573-6_16] [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] [Indexed: 06/15/2023]
Abstract
Drug addiction is a serious relapsing disease that has high costs to society and to the individual addicts. Treatment of these addictions is still in its nascency, with only a few examples of successful therapies. Therapeutic response depends upon genetic, biological, social, and environmental components. A role for genetic makeup in the response to treatment has been shown for several addiction pharmacotherapies with response to treatment based on individual genetic makeup. In this chapter, we will discuss the role of genetics in pharmacotherapies, specifically for cocaine, alcohol, and opioid dependences. The continued elucidation of the role of genetics should aid in the development of new treatments and increase the efficacy of existing treatments.
Collapse
Affiliation(s)
- David P Graham
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mark J Harding
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - David A Nielsen
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
18
|
Roos CR, Bold KW, Witkiewitz K, Leeman RF, DeMartini KS, Fucito LM, Corbin WR, Mann K, Kranzler HR, O’Malley SS. Reward drinking and naltrexone treatment response among young adult heavy drinkers. Addiction 2021; 116:2360-2371. [PMID: 33620746 PMCID: PMC8328878 DOI: 10.1111/add.15453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
AIMS Theory-driven, exploratory study to: (i) identify a reward drinking phenotype in young adults; (ii) evaluate this phenotype as a predictor of naltrexone response; and (iii) examine mechanisms of naltrexone in reward drinkers. DESIGN Secondary analysis of a randomized controlled trial. SETTING USA. PARTICIPANTS A total of 128 young adult (ages 18-25) heavy drinkers. INTERVENTIONS Naltrexone versus placebo. MEASUREMENTS Daily surveys assessed affect, urge, drinking, and context. The Drinking Motives Questionnaire was used to identify phenotypes based on reward (enhancement motives) and relief (coping motives) drinking. FINDINGS We identified three profiles: "Low reward/Low relief" (14.1%; low enhancement/low coping motives); "Reward drinkers" (62.2%; high enhancement/low coping motives); and "High reward/High relief" (22.7%; high enhancement/high coping motives). Among reward drinkers (versus low profile), naltrexone significantly reduced percent days drinking to intoxication (blood alcohol concentration [BAC] ≥0.08) (PDI) (d = 0.56; 95% CI [0.17, 0.96]) and percent high intensity drinking days (PHID) (8/10 drinks for women/men) (d = 0.32; 95% CI [0.01, 0.68]). Among the high reward/high relief profile drinkers (versus low profile), naltrexone reduced PHID (d = 0.69; 95% CI [0.02, 1.50]). Using profile-informed cutoffs and observed scores (for clinical applicability): (i) among cutoff-derived reward drinkers, we found a medium-to-large (d = 0.66; 95% CI [0.24, 1.16]) and small effect (d = 0.28; 95% CI [0.04, 0.72]) of naltrexone in reducing PDI and PHID, respectively; and (ii) among the cutoff-derived high reward/high relief subgroup, we found a medium-to-large effect (d = 0.63; 95% CI [0.05, 1.1]) of naltrexone in reducing PHID. Among reward drinkers (not other profiles), naltrexone reduced drinking on days a drinking event occurred by weakening the within-day association between positive affect and urges (P < 0.05). CONCLUSIONS Naltrexone has pronounced effects in reducing risky drinking among young adult reward drinkers (high reward/low relief) by reducing urges on days when individuals have higher positive affect and are exposed to a drinking event. Naltrexone also appears to reduce risky drinking among young adult high reward/high relief drinkers, but not via the same mechanism.
Collapse
Affiliation(s)
- Corey R. Roos
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | - Krysten W. Bold
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | - Katie Witkiewitz
- University of New Mexico, Department of Psychology, Albuquerque, NM
| | - Robert F. Leeman
- Yale School of Medicine, Department of Psychiatry, New Haven, CT,University of Florida, Department of Health Education and Behavior, Gainesville, FL
| | | | - Lisa M. Fucito
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | | | - Karl Mann
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim Germany
| | - Henry R. Kranzler
- Perelman School of Medicine, University of Pennsylvania and Crescenz VAMC, Philadelphia, PA
| | | |
Collapse
|
19
|
Ho MF, Zhang C, Zhang L, Wei L, Zhou Y, Moon I, Geske JR, Choi DS, Biernacka J, Frye M, Wen Z, Karpyak VM, Li H, Weinshilboum R. TSPAN5 influences serotonin and kynurenine: pharmacogenomic mechanisms related to alcohol use disorder and acamprosate treatment response. Mol Psychiatry 2021; 26:3122-3133. [PMID: 32753686 PMCID: PMC7858703 DOI: 10.1038/s41380-020-0855-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 01/13/2023]
Abstract
We previously reported that SNPs near TSPAN5 were associated with plasma serotonin (5-HT) concentrations which were themselves associated with selective serotonin reuptake inhibitor treatment outcomes in patients with major depressive disorder (MDD). TSPAN5 SNPs were also associated with alcohol consumption and alcohol use disorder (AUD) risk. The present study was designed to explore the biological function of TSPAN5 with a focus on 5-HT and kynurenine concentrations in the tryptophan pathway. Ethanol treatment resulted in decreased 5-HT concentrations in human induced pluripotent stem cell (iPSC)-derived neuron culture media, and the downregulation of gene expression of TSPAN5, DDC, MAOA, MAOB, TPH1, and TPH2 in those cells. Strikingly, similar observations were made when the cells were treated with acamprosate-an FDA approved drug for AUD therapy. These results were replicated in iPSC-derived astrocytes. Furthermore, TSPAN5 interacted physically with proteins related to clathrin and other vesicle-related proteins, raising the possibility that TSPAN5 might play a role in vesicular function in addition to regulating expression of genes associated with 5-HT biosynthesis and metabolism. Downregulation of TSPAN5 expression by ethanol or acamprosate treatment was also associated with decreased concentrations of kynurenine, a major metabolite of tryptophan that plays a role in neuroinflammation. Knockdown of TSPAN5 also influenced the expression of genes associated with interferon signaling pathways. Finally, we determined that TSPAN5 SNPs were associated with acamprosate treatment outcomes in AUD patients. In conclusion, TSPAN5 can modulate the concentrations of 5-HT and kynurenine. Our data also highlight a potentially novel pharmacogenomic mechanism related to response to acamprosate.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lingxin Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ying Zhou
- Department of Cell Biology, Emory University, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Irene Moon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jennifer R Geske
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Joanna Biernacka
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mark Frye
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Zhexing Wen
- Department of Cell Biology, Emory University, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
Domi E, Domi A, Adermark L, Heilig M, Augier E. Neurobiology of alcohol seeking behavior. J Neurochem 2021; 157:1585-1614. [PMID: 33704789 DOI: 10.1111/jnc.15343] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/29/2022]
Abstract
Alcohol addiction is a chronic relapsing brain disease characterized by an impaired ability to stop or control alcohol use despite adverse consequences. A main challenge of addiction treatment is to prevent relapse, which occurs in more than >50% of newly abstinent patients with alcohol disorder within 3 months. In people suffering from alcohol addiction, stressful events, drug-associated cues and contexts, or re-exposure to a small amount of alcohol trigger a chain of behaviors that frequently culminates in relapse. In this review, we first present the preclinical models that were developed for the study of alcohol seeking behavior, namely the reinstatement model of alcohol relapse and compulsive alcohol seeking under a chained schedule of reinforcement. We then provide an overview of the neurobiological findings obtained using these animal models, focusing on the role of opioids systems, corticotropin-release hormone and neurokinins, followed by dopaminergic, glutamatergic, and GABAergic neurotransmissions in alcohol seeking behavior.
Collapse
Affiliation(s)
- Esi Domi
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Heilig
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Eric Augier
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| |
Collapse
|
21
|
Borruto AM, Stopponi S, Li H, Weiss F, Roberto M, Ciccocioppo R. Genetically selected alcohol-preferring msP rats to study alcohol use disorder: Anything lost in translation? Neuropharmacology 2021; 186:108446. [PMID: 33476639 DOI: 10.1016/j.neuropharm.2020.108446] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022]
Abstract
For several decades, genetically selected alcohol-preferring rats have been successfully used to mimic and study alcohol use disorders (AUD). These rat lines have been instrumental in advancing our understanding of the neurobiology of alcoholism and enabling pharmacological studies to evaluate drug efficacy on alcohol drinking and relapse. Moreover, the results of these studies have identified genetic variables that are linked to AUD vulnerability. This is an up-to-date review that focuses on genetically selected Marchigian Sardinian alcohol-preferring (msP) rats. To support the translational relevance of the findings that are obtained from msP rats and highlight important similarities to AUD patients, we also discuss the results of recent brain imaging studies. Finally, to demonstrate the importance of studying sex differences in animal models of AUD, we present original data that highlight behavioral differences in the response to alcohol in male and female rats. Female msP rats exhibited higher alcohol consumption compared with males. Furthermore, msP rats of both sexes exhibit higher anxiety- and depressive-like behaviors in the elevated plus maze and forced swim test, respectively, compared with unselected Wistar controls. Notably, voluntary alcohol drinking decreases foot-shock stress and depressive-like behavior in both sexes, whereas anxiety-like behavior in the elevated plus maze is attenuated only in males. These findings suggest that male and female msP rats both drink high amounts of alcohol to self-medicate negative affective symptoms. For females, this behavior may be driven by an attempt to treat stress and depressive-like conditions. For males, generalized anxiety appears to be an important additional factor in the motivation to drink alcohol. This article is part of the special issue on 'Vulnerabilities to Substance Abuse.'
Collapse
Affiliation(s)
- Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Hongwu Li
- College of Chemical Engineering, Changchun University of Technology, Changchun, China
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| |
Collapse
|
22
|
Inoura S, Shimane T, Kitagaki K, Wada K, Matsumoto T. Parental drinking according to parental composition and adolescent binge drinking: findings from a nationwide high school survey in Japan. BMC Public Health 2020; 20:1878. [PMID: 33287791 PMCID: PMC7720457 DOI: 10.1186/s12889-020-09969-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Abstract
Background Alcohol problems in parents have been revealed to affect adolescent alcohol misuse. However, few studies examine the effects of parental drinking on adolescent risky drinking (including binge drinking) in the general population. In particular, previous study findings are inconsistent regarding the influence of parental drinking according to parental composition. In this study, we aimed to examine the relationship between parental drinking, according to parental composition, and binge drinking among high school students in Japan. Methods We performed a secondary analysis of the Nationwide High School Survey on Drug Use and Lifestyle 2018, Japan. A total of 46,848 valid surveys from high school students of 78 schools were included for analysis. Logistic regression analysis with a generalized linear mixed model was conducted with binge drinking as the dependent variable and “parental drinking according to parental composition” (e.g., father’s drinking, mother’s drinking, father’s absence, mother’s absence, both parents drinking, and neither parent at home) as the independent variable, after adjusting with covariates. Binge drinking was defined as five or more alcoholic drinks for male adolescents or four or more alcoholic drinks for females on the same occasion within two hours. Results In the fully adjusted models, adolescents whose mothers drink (adjusted odds ratio (AOR): 1.50, 95% confidence interval (CI): 1.06–2.12) were significantly associated with adolescent binge drinking. This risk was significantly higher among students with neither parent living at home (AOR: 4.35, 95% CI: 2.10–9.02). Conclusion Parental drinking and absence do affect adolescent binge drinking; our findings show that adolescents are more likely to engage in binge drinking if their mothers drink or if they are not living with either parent. Therefore, it is important to engage parents and non-parental family members in future programs and interventions to prevent adolescent binge drinking.
Collapse
Affiliation(s)
- Satoshi Inoura
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan.
| | - Takuya Shimane
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kunihiko Kitagaki
- Social Pharmacy Lab., Faculty of Pharmaceutical Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kiyoshi Wada
- Department of Addiction Treatment Research, Saitama Prefectural Psychiatric Hospital, Saitama, Japan
| | - Toshihiko Matsumoto
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
23
|
Padula AE, Rinker JA, Lopez MF, Mulligan MK, Williams RW, Becker HC, Mulholland PJ. Bioinformatics identification and pharmacological validation of Kcnn3/K Ca2 channels as a mediator of negative affective behaviors and excessive alcohol drinking in mice. Transl Psychiatry 2020; 10:414. [PMID: 33247097 PMCID: PMC7699620 DOI: 10.1038/s41398-020-01099-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/16/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Mood disorders are often comorbid with alcohol use disorder (AUD) and play a considerable role in the development and maintenance of alcohol dependence and relapse. Because of this high comorbidity, it is necessary to determine shared and unique genetic factors driving heavy drinking and negative affective behaviors. In order to identify novel pharmacogenetic targets, a bioinformatics analysis was used to quantify the expression of amygdala K+ channel genes that covary with anxiety-related phenotypes in the well-phenotyped and fully sequenced family of BXD strains. We used a model of stress-induced escalation of drinking in alcohol-dependent mice to measure negative affective behaviors during abstinence. A pharmacological approach was used to validate the key bioinformatics findings in alcohol-dependent, stressed mice. Amygdalar expression of Kcnn3 correlated significantly with 40 anxiety-associated phenotypes. Further examination of Kcnn3 expression revealed a strong eigentrait for anxiety-like behaviors and negative correlations with binge-like and voluntary alcohol drinking. Mice treated with chronic intermittent alcohol exposure and repeated swim stress consumed more alcohol in their home cages and showed hypophagia on the novelty-suppressed feeding test during abstinence. Pharmacologically targeting Kcnn gene products with the KCa2 (SK) channel-positive modulator 1-EBIO decreased drinking and reduced feeding latency in alcohol-dependent, stressed mice. Collectively, these validation studies provide central nervous system links into the covariance of stress, negative affective behaviors, and AUD in the BXD strains. Further, the bioinformatics discovery tool is effective in identifying promising targets (i.e., KCa2 channels) for treating alcohol dependence exacerbated by comorbid mood disorders.
Collapse
Affiliation(s)
- Audrey E Padula
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
24
|
Grigsby KB, Savarese AM, Metten P, Mason BJ, Blednov YA, Crabbe JC, Ozburn AR. Effects of Tacrolimus and Other Immune Targeting Compounds on Binge-Like Ethanol Drinking in High Drinking in the Dark Mice. Neurosci Insights 2020; 15:2633105520975412. [PMID: 33294845 PMCID: PMC7705291 DOI: 10.1177/2633105520975412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
High Drinking in the Dark (HDID-1) mice represent a unique genetic risk model of binge-like drinking and a novel means of screening potential pharmacotherapies to treat alcohol use disorders (AUDs). We tested the effects of tacrolimus (0, 0.5, 1, and 2 mg/kg), sirolimus (0, 5, 10, and 20 mg/kg), palmitoylethanolamide (PEA; 0, 75, 150, and 225 mg/kg), and secukinumab (0, 5, 20, and 60 mg/kg) on binge-like ethanol intake (2-day, "Drinking in the Dark" [DID]) and blood alcohol levels (BALs) in HDID-1 mice. Tacrolimus reduced ethanol intake and BALs. Tacrolimus had no effect on water intake, but reduced saccharin intake. There was no effect of sirolimus, PEA, or secukinumab on ethanol intake or BALs. These results compare and contrast with previous work addressing these compounds or their targeted mechanisms of action on ethanol drinking, highlighting the importance of screening a wide range of models and genotypes to inform the role of neuroimmune signaling in AUDs.
Collapse
Affiliation(s)
- Kolter B Grigsby
- Portland Alcohol Research Center,
Department of Behavioral Neuroscience at Oregon Health and Science University and VA
Portland Health Care System, Portland, OR, USA
| | - Antonia M Savarese
- Portland Alcohol Research Center,
Department of Behavioral Neuroscience at Oregon Health and Science University and VA
Portland Health Care System, Portland, OR, USA
| | - Pamela Metten
- Portland Alcohol Research Center,
Department of Behavioral Neuroscience at Oregon Health and Science University and VA
Portland Health Care System, Portland, OR, USA
| | - Barbara J Mason
- Department of Molecular Medicine, The
Scripps Research Institute, La Jolla, CA, USA
| | - Yuri A Blednov
- Waggoner Center for Alcoholism and
Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - John C Crabbe
- Portland Alcohol Research Center,
Department of Behavioral Neuroscience at Oregon Health and Science University and VA
Portland Health Care System, Portland, OR, USA
| | - Angela R Ozburn
- Portland Alcohol Research Center,
Department of Behavioral Neuroscience at Oregon Health and Science University and VA
Portland Health Care System, Portland, OR, USA
| |
Collapse
|
25
|
Neuroimaging reveals functionally distinct neuronal networks associated with high-level alcohol consumption in two genetic rat models. Behav Pharmacol 2020; 32:229-238. [PMID: 32925226 DOI: 10.1097/fbp.0000000000000582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human imaging data suggest that the motivational processes associated with alcohol reward are reflected in the patterns of neural activation after alcohol or alcohol-related cues. In animal models of alcohol drinking, however, the changes in brain activation during voluntary alcohol ingestion are poorly known. In order to improve the translational utility of animal models, we examined alcohol-induced functional brain activation in Alko Alcohol (AA) and Marchigian-Sardinian alcohol-preferring (msP) rats that drink voluntarily high levels of alcohol, but exhibit widely different neurochemical and behavioral traits cosegregated with alcohol preference. Brain imaging was performed using manganese-enhanced MRI (MEMRI), which is based on accumulation of Mn2+ ions in activated neurons, allowing the identification of functional neuronal networks recruited during specific behaviors in awake animals during a subsequent imaging session under anesthesia. MEMRI was performed following 4 weeks of voluntary alcohol drinking, using water drinking as the control. Despite similar levels of alcohol drinking, strikingly different alcohol-induced neuronal activity patterns were observed in AA and msP rats. Overall, functional activation in the AA rats was more widespread, involving large cortical areas and subcortical structures, such as the bed nucleus of the stria terminalis, preoptic area, hypothalamus, periaqueductal grey, and substantia nigra. In the msP rats, however, alcohol-related activation was largely confined to prefrontal cortical regions and insular cortex, and olfactory areas. Overlapping areas of activation found in both rat lines included the nucleus accumbens, prelimbic, orbital, and insular cortex. In conclusion, our data reveal strikingly different brain circuits associated with alcohol drinking in two genetically different rat lines and suggest innately different motivational and behavioral processes driving alcohol drinking. These findings have important implications for the use of these lines in translational alcohol research.
Collapse
|
26
|
Suárez J, Khom S, Alén F, Natividad LA, Varodayan FP, Patel RR, Kirson D, Arco R, Ballesta A, Bajo M, Rubio L, Martin-Fardon R, de Fonseca FR, Roberto M. Cessation of fluoxetine treatment increases alcohol seeking during relapse and dysregulates endocannabinoid and glutamatergic signaling in the central amygdala. Addict Biol 2020; 25:e12813. [PMID: 31339221 PMCID: PMC8050940 DOI: 10.1111/adb.12813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023]
Abstract
Administration of selective serotonin reuptake inhibitors (SSRIs), typically used as antidepressants, induces long-lasting behavioral changes associated with alcohol use disorder (AUD). However, the contribution of SSRI (fluoxetine)-induced alterations in neurobiological processes underlying alcohol relapse such as endocannabinoid and glutamate signaling in the central amygdala (CeA) remains largely unknown. We utilized an integrative approach to study the effects of repeated fluoxetine administration during abstinence on ethanol drinking. Gene expression and biochemical and electrophysiological studies explored the hypothesis that dysregulation in glutamatergic and endocannabinoid mechanisms in the CeA underlie the susceptibility to alcohol relapse. Cessation of daily treatment with fluoxetine (10 mg/kg) during abstinence resulted in a marked increase in ethanol seeking during re-exposure periods. The increase in ethanol self-administration was associated with (a) reductions in levels of the endocannabinoids N-arachidonoylethanolomine and 2-arachidonoylglycerol in the CeA, (b) increased amygdalar gene expression of cannabinoid type-1 receptor (CB1), N-acyl phosphatidylethanolamine phospholipase D (Nape-pld), fatty acid amid hydrolase (Faah), (c) decreased amygdalar gene expression of ionotropic AMPA (GluA2 and GluA4) and metabotropic (mGlu3) glutamate receptors, and (d) increased glutamatergic receptor function. Overall, our data suggest that the administration of the antidepressant fluoxetine during abstinence dysregulates endocannabinoid signaling and glutamatergic receptor function in the amygdala, facts that likely facilitate alcohol drinking behavior during relapse.
Collapse
Affiliation(s)
- Juan Suárez
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Fulbright Visiting Scholar Program, Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Sophia Khom
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Francisco Alén
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Luis A. Natividad
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Florence P. Varodayan
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Reesha R. Patel
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Dean Kirson
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Rocío Arco
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
| | - Antonio Ballesta
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Michal Bajo
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Leticia Rubio
- Department of Anatomy and Forensic and Legal Medicine. Universidad de Málaga, Málaga, Spain
| | - Rémi Martin-Fardon
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Malaga (IBIMA), Mental Health UGC, Hospital Universitario Regional de Málaga, Málaga, Spain
- Department of Psychobiology. Universidad Complutense de Madrid, Madrid, Spain
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute (TSRI), La Jolla, CA, USA
| |
Collapse
|
27
|
Wei Y, Liu L, Su X, Zhao L, Jiang H. Precision medicine: Subgroup identification in longitudinal trajectories. Stat Methods Med Res 2020; 29:2603-2616. [PMID: 32070237 PMCID: PMC8357634 DOI: 10.1177/0962280220904114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In clinical studies, the treatment effect may be heterogeneous among patients. It is of interest to identify subpopulations which benefit most from the treatment, regardless of the treatment's overall performance. In this study, we are interested in subgroup identification in longitudinal studies when nonlinear trajectory patterns are present. Under such a situation, evaluation of the treatment effect entails comparing longitudinal trajectories while subgroup identification requires a further evaluation of differential treatment effects among subgroups induced by moderators. To this end, we propose a tree-structured subgroup identification method, termed "interaction tree for longitudinal trajectories", which combines mixed effects models with regression splines to model the nonlinear progression patterns among repeated measures. Extensive simulation studies are conducted to evaluate its performance and an application to an alcohol addiction pharmacogenetic trial is presented.
Collapse
Affiliation(s)
- Yishu Wei
- Department of statistics, Northwestern University, Evanston, IL, United States
| | - Lei Liu
- Division of biostatistics, Washington University, St. Louis, MO, United States
| | - Xiaogang Su
- Department of Mathematical Sciences, University of Texas at El Paso (UTEP), El Paso, TX, United States
| | - Lihui Zhao
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Hongmei Jiang
- Department of statistics, Northwestern University, Evanston, IL, United States
| |
Collapse
|
28
|
Lieberman R, Jensen KP, Clinton K, Levine ES, Kranzler HR, Covault J. Molecular Correlates of Topiramate and GRIK1 rs2832407 Genotype in Pluripotent Stem Cell-Derived Neural Cultures. Alcohol Clin Exp Res 2020; 44:1561-1570. [PMID: 32574382 PMCID: PMC7491603 DOI: 10.1111/acer.14399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND There is growing evidence that the anticonvulsant topiramate is efficacious in reducing alcohol consumption. Further, an intronic single nucleotide polymorphism (rs2832407, C A) in the GRIK1 gene, which encodes the GluK1 subunit of the excitatory kainate receptor, predicted topiramate's effectiveness in reducing heavy drinking in a clinical trial. The molecular correlates of GRIK1 genotype that may relate to topiramate's ability to reduce drinking remain unknown. METHODS We differentiated induced pluripotent stem cells (iPSCs) characterized by GRIK1 rs2832407 genotype from 8 A/A and 8 C/C donors into forebrain-lineage neural cultures. Our differentiation protocol yielded mixed neural cultures enriched for glutamatergic neurons. Basal mRNA expression of the GRIK1 locus was examined via quantitative polymerase chain reaction (qPCR). The effects of acute topiramate exposure on excitatory spontaneous synaptic activity were examined via whole-cell patch-clamp electrophysiology. Results were compared and contrasted between iPSC donor genotypes. RESULTS Although characterization of the GRIK1 locus revealed no effect of rs2832407 genotype on GRIK1 isoform mRNA expression, a significant difference was observed on GRIK1 antisense-2 expression, which was greater in C/C neural cultures. Differential effects of acute exposure to 5 μM topiramate were observed on spontaneous synaptic activity in A/A versus C/C neurons, with a smaller reduction in excitatory event frequency observed in C/C donor neurons. CONCLUSIONS This work highlights the use of iPSC technologies to study pharmacogenetic treatment effects in psychiatric disorders and furthers our understanding of the molecular effects of topiramate exposure in human neural cells.
Collapse
Affiliation(s)
- Richard Lieberman
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06030-1410
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA 06030
| | - Kevin P. Jensen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511
- VA Connecticut Healthcare System, West Haven, CT 06516
| | - Kaitlin Clinton
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06030-1410
| | - Eric S. Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA 06030
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104
- VISN4 MIRECC, Crescenz VAMC, Philadelphia, PA 19104
| | - Jonathan Covault
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06030-1410
| |
Collapse
|
29
|
Wu R, Liu J, Wang K, Huang Y, Zhang Y, Li JX. Effects of a trace amine-associated receptor 1 agonist RO 5263397 on ethanol-induced behavioral sensitization. Behav Brain Res 2020; 390:112641. [PMID: 32407821 DOI: 10.1016/j.bbr.2020.112641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alcohol dependence is a chronic and severe health problem which puts a heavy burden on society. Alcohol activates mesolimbic dopamine circuity to achieve its reinforcing effect. While TAAR1 is critically involved in the modulation of dopamine, there is little evidence indicating that TAAR1 could play a role in behavioral effects of ethanol. METHODS By using the animal model of behavioral sensitization induced by ethanol in mice, the present study was performed to investigate whether the activation of TAAR1 would affect the behavioral plasticity of ethanol. RESULTS Repeated administration with ethanol induced a significant increased locomotion in WT mice with females showing higher level of sensitization to ethanol than male mice. The TAAR1 agonist RO5263397 significantly decreased the expression of ethanol-induced behavioral sensitization both in male and female WT mice (0.1 and 0.32 mg/kg). Repeated RO5263397 exposure also prevented the development of behavioral sensitization to ethanol both in male and female WT mice. Moreover, while TAAR1-KO mice developed normal levels of ethanol-induced behavioral sensitization, RO5263397 did not affect this behavior in TAAR1-KO mice. CONCLUSIONS These results indicated that the TAAR1 agonist RO5263397 negatively regulated the expression and development of ethanol-elicited behavioral sensitization in WT but not in TAAR1-KO mice. The present study suggests that TAAR1 is probably involved in certain addiction-like effects of alcohol and could be a useful drug target for the development of new medications to treat alcohol dependence.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States; School of Medicine, Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Kaixuan Wang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yufei Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yanan Zhang
- Research Triangle Institute, NC, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
30
|
Peterson VL, Richards JB, Meyer PJ, Cabrera-Rubio R, Tripi JA, King CP, Polesskaya O, Baud A, Chitre AS, Bastiaanssen TFS, Woods LS, Crispie F, Dinan TG, Cotter PD, Palmer AA, Cryan JF. Sex-dependent associations between addiction-related behaviors and the microbiome in outbred rats. EBioMedicine 2020; 55:102769. [PMID: 32403084 PMCID: PMC7218262 DOI: 10.1016/j.ebiom.2020.102769] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Multiple factors contribute to the etiology of addiction, including genetics, sex, and a number of addiction-related behavioral traits. One behavioral trait where individuals assign incentive salience to food stimuli ("sign-trackers", ST) are more impulsive compared to those that do not ("goal-trackers", GT), as well as more sensitive to drugs and drug stimuli. Furthermore, this GT/ST phenotype predicts differences in other behavioral measures. Recent studies have implicated the gut microbiota as a key regulator of brain and behavior, and have shown that many microbiota-associated changes occur in a sex-dependent manner. However, few studies have examined how the microbiome might influence addiction-related behaviors. To this end, we sought to determine if gut microbiome composition was correlated with addiction-related behaviors determined by the GT/ST phenotype. METHODS Outbred male (N=101) and female (N=101) heterogeneous stock rats underwent a series of behavioral tests measuring impulsivity, attention, reward-learning, incentive salience, and locomotor response. Cecal microbiome composition was estimated using 16S rRNA gene amplicon sequencing. Behavior and microbiome were characterized and correlated with behavioral phenotypes. Robust sex differences were observed in both behavior and microbiome; further analyses were conducted within sex using the pre-established goal/sign-tracking (GT/ST) phenotype and partial least squares differential analysis (PLS-DA) clustered behavioral phenotype. RESULTS Overall microbiome composition was not associated to the GT/ST phenotype. However, microbial alpha diversity was significantly decreased in female STs. On the other hand, a measure of impulsivity had many significant correlations to microbiome in both males and females. Several measures of impulsivity were correlated with the genus Barnesiella in females. Female STs had notable correlations between microbiome and attentional deficient. In both males and females, many measures were correlated with the bacterial families Ruminocococcaceae and Lachnospiraceae. CONCLUSIONS These data demonstrate correlations between several addiction-related behaviors and the microbiome specific to sex.
Collapse
Affiliation(s)
- Veronica L Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Room 2.33, 2nd Floor, Western Gateway Building, Cork, Ireland
| | - Jerry B Richards
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Paul J Meyer
- Department of Psychology, University at Buffalo, Buffalo, NY, USA
| | - Raul Cabrera-Rubio
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Jordan A Tripi
- Department of Psychology, University at Buffalo, Buffalo, NY, USA
| | | | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, CA, USA
| | - Amelie Baud
- Department of Psychiatry, University of California San Diego, CA, USA
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, CA, USA
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Room 2.33, 2nd Floor, Western Gateway Building, Cork, Ireland
| | | | - Fiona Crispie
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, CA, USA; Institute for Genomic Medicine, University of California San Diego, CA, USA; Center for Microbiome Innovation, University of California San Diego, CA, USA
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Room 2.33, 2nd Floor, Western Gateway Building, Cork, Ireland.
| |
Collapse
|
31
|
Hood LE, Leyrer-Jackson JM, Olive MF. Pharmacotherapeutic management of co-morbid alcohol and opioid use. Expert Opin Pharmacother 2020; 21:823-839. [PMID: 32103695 PMCID: PMC7239727 DOI: 10.1080/14656566.2020.1732349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022]
Abstract
Opioid use disorder (OUD) and alcohol use disorder (AUD) are two highly prevalent substance-related disorders worldwide. Co-use of the substances is also quite prevalent, yet there are no pharmacological treatment approaches specifically designed to treat co-morbid OUD and AUD. Here, the authors critically summarize OUD, AUD and opioid/alcohol co-use and their current pharmacotherapies for treatment. They also review the mechanisms of action of opioids and alcohol within the brain reward circuitry and discuss potential combined mechanisms of action and resulting neuroadaptations. Pharmacotherapies that aim to treat AUD or OUD that may be beneficial in the treatment of co-use are also highlighted. Preclinical models assessing alcohol and opioid co-use remain sparse. Lasting neuroadaptations in brain reward circuits caused by co-use of alcohol and opioids remains largely understudied. In order to fully understand the neurobiological underpinnings of alcohol and opioid co-use and develop efficacious pharmacotherapies, the preclinical field must expand its current experimental paradigms of 'single drug' use to encompass polysubstance use. Such studies will provide insights on the neural alterations induced by opioid and alcohol co-use, and may help develop novel pharmacotherapies for individuals with co-occurring alcohol and opioid use disorders.
Collapse
Affiliation(s)
- Lauren E. Hood
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | | | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
32
|
Haass-Koffler CL. Protein Tyrosine Phosphatase β/ζ and Alcohol Use Disorder: A Commentary. Alcohol Clin Exp Res 2020; 44:1189-1191. [PMID: 32343842 DOI: 10.1111/acer.14346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Carolina L Haass-Koffler
- From the, Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island.,Department of Psychiatry and Human Behavior, Werren Alpert Medical School, Brown University, Providence, Rhode Island.,Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island
| |
Collapse
|
33
|
Voon V, Grodin E, Mandali A, Morris L, Doñamayor N, Weidacker K, Kwako L, Goldman D, Koob GF, Momenan R. Addictions NeuroImaging Assessment (ANIA): Towards an integrative framework for alcohol use disorder. Neurosci Biobehav Rev 2020; 113:492-506. [PMID: 32298710 DOI: 10.1016/j.neubiorev.2020.04.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/30/2020] [Accepted: 04/05/2020] [Indexed: 02/08/2023]
Abstract
Alcohol misuse and addiction are major international public health issues. Addiction can be characterized as a disorder of aberrant neurocircuitry interacting with environmental, genetic and social factors. Neuroimaging in alcohol misuse can thus provide a critical window into underlying neural mechanisms, highlighting possible treatment targets and acting as clinical biomarkers for predicting risk and treatment outcomes. This neuroimaging review on alcohol misuse in humans follows the Addictions Neuroclinical Assessment (ANA) that proposes incorporating three functional neuroscience domains integral to the neurocircuitry of addiction: incentive salience and habits, negative emotional states, and executive function within the context of the addiction cycle. Here we review and integrate multiple imaging modalities focusing on underlying cognitive processes such as reward anticipation, negative emotionality, cue reactivity, impulsivity, compulsivity and executive function. We highlight limitations in the literature and propose a model forward in the use of neuroimaging as a tool to understanding underlying mechanisms and potential clinical applicability for phenotyping of heterogeneity and predicting risk and treatment outcomes.
Collapse
Affiliation(s)
- Valerie Voon
- Department of Psychiatry, University of Cambridge, Cambridge, UK; Behavioural and Clinical Neurosciences Institute, Cambridge, UK; Cambridgeshire and Peterborough NHS Trust, Cambridge, UK.
| | - Erica Grodin
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, UK
| | - Alekhya Mandali
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Laurel Morris
- Behavioural and Clinical Neurosciences Institute, Cambridge, UK; Department of Psychology, University of Cambridge, Cambridge, UK
| | - Nuria Doñamayor
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | - Laura Kwako
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, UK
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, UK
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, UK
| | - Reza Momenan
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, UK
| |
Collapse
|
34
|
Song S, Marcum CS, Wilkinson AV, Shete S, Koehly LM. Genetic, Psychological, and Personal Network Factors Associated With Changes in Binge Drinking Over 2 Years Among Mexican Heritage Adolescents in the USA. Ann Behav Med 2020; 53:126-137. [PMID: 29697747 DOI: 10.1093/abm/kay019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Despite prevalent binge drinking and alcohol-dependent symptoms among Hispanics, few studies have examined how multidimensional factors influence Hispanic adolescents' binge drinking. Purpose This study examines the effects of genetic, psychological, and social network factors on binge drinking over time among Mexican heritage adolescents in the USA and whether there are correlations among genetic variants that are associated with binge drinking and psychological and network characteristics. Methods Mexican heritage adolescents (n = 731) participated in a longitudinal study, which included genetic testing at baseline, alcohol use assessments at first and second follow-ups, and questionnaires on sensation seeking, impulsivity, and peer and family network characteristics at second follow-up. Logistic regression and Spearman correlation analyses were performed. Results After adjusting for demographic characteristics, underlying genetic clustering, and binge drinking at first follow-up, two genetic variants on tryptophan hydroxylase 2 (TPH2; rs17110451, rs7963717), sensation seeking and impulsivity, and having a greater fraction of peers who drink or encourage drinking alcohol were associated with greater risk whereas another genetic variant on TPH2 (rs11178999) and having a greater fraction of close family relationships were associated with reduced risk for binge drinking at second follow-up. Genetic variants in TPH1 (rs591556) were associated with sensation seeking and impulsivity, while genetic variants in TPH2 (rs17110451) were associated with the fraction of drinkers in family. Conclusions Results reveal that genetic variants in the serotonin pathway, behavioral disinhibition traits, and social networks exert joint influences on binge drinking in Mexican heritage adolescents in the USA.
Collapse
Affiliation(s)
- Sunmi Song
- Department of Healthy Living, Health Risk Prevention Team, Korea Health Promotion Institute, Jung-gu, Seoul, Republic of Korea
| | - Christopher Steven Marcum
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna V Wilkinson
- Department of Epidemiology, Human Genetics and Environmental Science, UTHealth School of Public Health in Austin, Austin, Texas, USA
| | - Sanjay Shete
- Division of Quantitative Sciences, University of Texas MD Anderson Center, Houston, Texas, USA
| | - Laura M Koehly
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Bahi A, Dreyer JL. Environmental enrichment decreases chronic psychosocial stress-impaired extinction and reinstatement of ethanol conditioned place preference in C57BL/6 male mice. Psychopharmacology (Berl) 2020; 237:707-721. [PMID: 31786650 DOI: 10.1007/s00213-019-05408-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022]
Abstract
RATIONALE During the last few decades, alcohol use disorders (AUD) have reached an epidemic prevalence, yet social influences on alcoholism have not been fully addressed. Several factors can modulate alcohol intake. On one hand, stress can reinforce ethanol-induced behaviors and be an important component in AUD and alcoholism. On the other hand, environmental enrichment (EE) has a neuroprotective role and prevents the development of excessive ethanol intake in rodents. However, studies showing the role of EE in chronic psychosocial stress-impaired ethanol-conditioned rewards are nonexistent. AIM The purpose of the current study is to explore the potential protective role of EE on extinction and reinstatement of ethanol-conditioned place preference (EtOH-CPP) following chronic psychosocial stress. METHODS In the first experiment and after the EtOH-CPP test, the mice were subjected to 15 days of chronic stress, then housed in a standard (SE) or enriched environment (EE) while EtOH-CPP extinction was achieved by repeated exposure to the CPP chambers without ethanol injection. In the second experiment and after the EtOH-CPP test, extinction was achieved as described above. Mice were then exposed to chronic stress for 2 weeks before being housed in a SE or EE. EtOH-CPP reinstatement was induced by a single exposure to the conditioning chambers. RESULTS As expected, stress exposure increased anxiety-like behavior and reduced weight gain. More importantly, we found that EE significantly shortened chronic stress-delayed extinction and decreased the reinstatement of EtOH-CPP. CONCLUSION These results support the hypothesis that EE reduces the impact of alcohol-associated environmental stimuli, and hence it may be a general intervention for reducing cue-elicited craving and relapse in humans.
Collapse
Affiliation(s)
- Amine Bahi
- College of Medicine, Ajman University, Ajman, UAE. .,Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE.
| | - Jean-Luc Dreyer
- Division of Biochemistry, Department of Medicine, University of Fribourg, 1700, Fribourg, Switzerland
| |
Collapse
|
36
|
Hendershot CS, Dermody SS, Wardell JD, Zaso MJ, Kennedy JL, Stoner SA. OPRM1 Moderates Daily Associations of Naltrexone Adherence With Alcohol Consumption: Preliminary Evidence From a Mobile Health Trial. Alcohol Clin Exp Res 2020; 44:983-991. [PMID: 32020635 DOI: 10.1111/acer.14300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/25/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Initial evidence that OPRM1 genotype moderates the clinical response to naltrexone has not been replicated in prospective clinical trials. However, the use of traditional statistical analyses and clinical endpoints might limit sensitivity for studying pharmacogenetic associations, whereas the use of intensive daily assessments and person-centered analytic methods might increase sensitivity. This study leveraged person-centered analyses and daily measures of alcohol use, craving, and medication adherence to investigate OPRM1 as a moderator of changes in clinical outcomes during naltrexone treatment. METHODS Treatment-seeking participants with alcohol use disorder (n = 58; Mage = 38 years; 71% male) provided daily cell phone reports of craving and consumption while taking naltrexone as part of a mobile health trial. Daily medication adherence was measured remotely using electronic pill cap recordings. Multilevel modeling and multilevel structural equation modeling analyses evaluated the hypotheses that OPRM1 genotype would moderate prospective reductions in daily alcohol use and craving, and would also moderate within-person associations of daily adherence with same-day craving and consumption. RESULTS OPRM1 genotype moderated the association of daily adherence with reduced same-day consumption (p = 0.007) and craving (p = 0.06), with these associations being stronger for participants with the 118G variant. OPRM1 genotype did not moderate changes in craving and consumption over time. CONCLUSIONS These findings suggest that high-density assessments and person-centered analytic approaches, including modeling within-person variation in medication adherence, could be advantageous for pharmacogenetic studies.
Collapse
Affiliation(s)
- Christian S Hendershot
- From the, Campbell Family Mental Health Research Institute, (CSH, JDW, JLK), Centre for Addiction and Mental Health, Toronto, ON, Canada.,Institute for Mental Health Policy Research, (CSH, JDW), Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, (CSH, JDW, JLK), University of Toronto, Toronto, ON, Canada.,Department of Psychology, (CSH), University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, (CSH), University of Toronto, Toronto, ON, Canada
| | - Sarah S Dermody
- School of Psychological Science, (SSD), Oregon State University, Corvallis, Oregon
| | - Jeffrey D Wardell
- From the, Campbell Family Mental Health Research Institute, (CSH, JDW, JLK), Centre for Addiction and Mental Health, Toronto, ON, Canada.,Institute for Mental Health Policy Research, (CSH, JDW), Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, (CSH, JDW, JLK), University of Toronto, Toronto, ON, Canada
| | - Michelle J Zaso
- Clinical and Research Institute on Addictions, (MJZ), University at Buffalo, Buffalo, New York
| | - James L Kennedy
- From the, Campbell Family Mental Health Research Institute, (CSH, JDW, JLK), Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, (CSH, JDW, JLK), University of Toronto, Toronto, ON, Canada
| | - Susan A Stoner
- Alcohol and Drug Abuse Institute, (SAS), University of Washington, Seattle, Washington
| |
Collapse
|
37
|
Regier PS, Kampman KM, Childress AR. Clinical Trials for Stimulant Use Disorders: Addressing Heterogeneities That May Undermine Treatment Outcomes. Handb Exp Pharmacol 2020; 258:299-322. [PMID: 32193666 DOI: 10.1007/164_2019_303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In recent years, use of cocaine and amphetamines and deaths associated with stimulants have been on the rise, and there are still no FDA-approved medications for stimulant use disorders. One contributing factor may involve heterogeneity. At the neurobiological level, dual dopamine dysfunction may be undermining medication efficacy, suggesting a need for combination pharmacotherapies. At the population level, individual variability is expressed in a number of ways and, if left unaddressed, may interfere with medication efficacy. This chapter reviews studies investigating medications to address dopamine dysfunction, and it also identifies several prominent heterogeneities associated with stimulant (and other substance) use disorders. The chapter has implications for improving interventions to treat stimulant use disorders, and the theme of individual heterogeneity may have broader application across substance use disorders.
Collapse
Affiliation(s)
- Paul S Regier
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA.
| | - Kyle M Kampman
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Rose Childress
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Hansson AC, Gründer G, Hirth N, Noori HR, Spanagel R, Sommer WH. Dopamine and opioid systems adaptation in alcoholism revisited: Convergent evidence from positron emission tomography and postmortem studies. Neurosci Biobehav Rev 2019; 106:141-164. [DOI: 10.1016/j.neubiorev.2018.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/08/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
|
39
|
Translational Molecular Approaches in Substance Abuse Research. Handb Exp Pharmacol 2019; 258:31-60. [PMID: 31628598 DOI: 10.1007/164_2019_259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Excessive abuse of psychoactive substances is one of the leading contributors to morbidity and mortality worldwide. In this book chapter, we review translational research strategies that are applied in the pursuit of new and more effective therapeutics for substance use disorder (SUD). The complex, multidimensional nature of psychiatric disorders like SUD presents difficult challenges to investigators. While animal models are critical for outlining the mechanistic relationships between defined behaviors and genetic and/or molecular changes, the heterogeneous pathophysiology of brain diseases is uniquely human, necessitating the use of human studies and translational research schemes. Translational research describes a cross-species approach in which findings from human patient-based data can be used to guide molecular genetic investigations in preclinical animal models in order to delineate the mechanisms of reward circuitry changes in the addicted state. Results from animal studies can then inform clinical investigations toward the development of novel treatments for SUD. Here we describe the strategies that are used to identify and functionally validate genetic variants in the human genome which may contribute to increased risk for SUD, starting from early candidate gene approaches to more recent genome-wide association studies. We will next examine studies aimed at understanding how transcriptional and epigenetic dysregulation in SUD can persistently alter cellular function in the disease state. In our discussion, we then focus on examples from the literature illustrating molecular genetic methodologies that have been applied to studies of different substances of abuse - from alcohol and nicotine to stimulants and opioids - in order to exemplify how these approaches can both delineate the underlying molecular systems driving drug addiction and provide insights into the genetic basis of SUD.
Collapse
|
40
|
Gomis-Vicent E, Thoma V, Turner JJD, Hill KP, Pascual-Leone A. Review: Non-Invasive Brain Stimulation in Behavioral Addictions: Insights from Direct Comparisons With Substance Use Disorders. Am J Addict 2019; 28:431-454. [PMID: 31513324 DOI: 10.1111/ajad.12945] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/01/2019] [Accepted: 07/27/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Treatment models developed for substance use disorders (SUDs) are often applied to behavioral addictions (BAs), even though the correspondence between these forms of addiction is unclear. This is also the case for noninvasive brain stimulation (NIBS) techniques being investigated as potential treatment interventions for SUDs and BAs. OBJECTIVES to contribute to the development of more effective NIBS protocols for BAs. METHODS Two literature searches using PubMed and Google Scholar were conducted identifying a total of 35 studies. The first search identified 25 studies examining the cognitive and neurophysiological overlap between BAs and SUDs. The second search yielded 10 studies examining the effects of NIBS in BAs. RESULTS Impulsivity and cravings show behavioral and neurophysiologic overlaps between BAs and SUDs, however, other outcomes like working-memory abilities or striatal connectivity, differ between BAs and SUDs. The most-employed NIBS target in BAs was dorsolateral prefrontal cortex (DLPFC), which was associated with a decrease in cravings, and less frequently with a reduction of addiction severity. CONCLUSIONS AND SCIENTIFIC SIGNIFICANCE Direct comparisons between BAs and SUDs revealed discrepancies between behavioral and neurophysiological outcomes, but overall, common and distinctive characteristics underlying each disorder. The lack of complete overlap between BAs and SUDs suggests that investigating the cognitive and neurophysiological features of BAs to create individual NIBS protocols that target risk-factors associated specifically with BAs, might be more effective than transferring protocols from SUDs to BAs. Individualizing NIBS protocols to target specific risk-factors associated with each BA might help to improve treatment interventions for BAs. (Am J Addict 2019;00:1-23).
Collapse
Affiliation(s)
- Elena Gomis-Vicent
- Department of Psychological Sciences, College of Applied Health and Communities, University of East London, London, United Kingdom
| | - Volker Thoma
- Department of Psychological Sciences, College of Applied Health and Communities, University of East London, London, United Kingdom
| | - John J D Turner
- Department of Psychological Sciences, College of Applied Health and Communities, University of East London, London, United Kingdom
| | - Kevin P Hill
- Division of Addiction Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Alvaro Pascual-Leone
- Department of Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Institut Guttmann de Neurorehabilitació, Universitat Autonòma de Barcelona, Badalona, Barcelona, Spain
| |
Collapse
|
41
|
Melbourne JK, Thompson KR, Peng H, Nixon K. Its complicated: The relationship between alcohol and microglia in the search for novel pharmacotherapeutic targets for alcohol use disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:179-221. [PMID: 31601404 DOI: 10.1016/bs.pmbts.2019.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disorder with wide-ranging health consequences. Alcohol targets the central nervous system producing neurodegeneration and subsequent cognitive and behavioral deficits, but the mechanisms behind these effects remain unclear. Recently, evidence has been mounting for the role of neuroimmune activation in the pathogenesis of AUDs, but our nascent state of knowledge about the interaction of alcohol with the neuroimmune system supports that the relationship is complicated. As the resident macrophage of the central nervous system, microglia are a central focus. Human and animal research on the interplay between microglia and alcohol in AUDs has proven to be complex, and though early research focused on a pro-inflammatory phenotype of microglia, the anti-inflammatory and homeostatic roles of microglia must be considered. How these new roles for microglia should be incorporated into our thinking about the neuroimmune system in AUDs is discussed in the context of developing novel pharmacotherapies for AUDs.
Collapse
Affiliation(s)
- Jennifer K Melbourne
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - K Ryan Thompson
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - Hui Peng
- University of Kentucky, College of Pharmacy, Department of Pharmaceutical Sciences, Lexington, KY, United States
| | - Kimberly Nixon
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States.
| |
Collapse
|
42
|
Molecular, Morphological, and Functional Characterization of Corticotropin-Releasing Factor Receptor 1-Expressing Neurons in the Central Nucleus of the Amygdala. eNeuro 2019; 6:ENEURO.0087-19.2019. [PMID: 31167849 PMCID: PMC6584068 DOI: 10.1523/eneuro.0087-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/12/2019] [Accepted: 05/25/2019] [Indexed: 01/28/2023] Open
Abstract
The central nucleus of the amygdala (CeA) is a brain region implicated in anxiety, stress-related disorders and the reinforcing effects of drugs of abuse. Corticotropin-releasing factor (CRF, Crh) acting at cognate type 1 receptors (CRF1, Crhr1) modulates inhibitory and excitatory synaptic transmission in the CeA. Here, we used CRF1:GFP reporter mice to characterize the morphological, neurochemical and electrophysiological properties of CRF1-expressing (CRF1+) and CRF1-non-expressing (CRF1-) neurons in the CeA. We assessed these two neuronal populations for distinctions in the expression of GABAergic subpopulation markers and neuropeptides, dendritic spine density and morphology, and excitatory transmission. We observed that CeA CRF1+ neurons are GABAergic but do not segregate with calbindin (CB), calretinin (CR), parvalbumin (PV), or protein kinase C-δ (PKCδ). Among the neuropeptides analyzed, Penk and Sst had the highest percentage of co-expression with Crhr1 in both the medial and lateral CeA subdivisions. Additionally, CeA CRF1+ neurons had a lower density of dendritic spines, which was offset by a higher proportion of mature spines compared to neighboring CRF1- neurons. Accordingly, there was no difference in basal spontaneous glutamatergic transmission between the two populations. Application of CRF increased overall vesicular glutamate release onto both CRF1+ and CRF1- neurons and does not affect amplitude or kinetics of EPSCs in either population. These novel data highlight important differences in the neurochemical make-up and morphology of CRF1+ compared to CRF1- neurons, which may have important implications for the transduction of CRF signaling in the CeA.
Collapse
|
43
|
Cannella N, Ubaldi M, Masi A, Bramucci M, Roberto M, Bifone A, Ciccocioppo R. Building better strategies to develop new medications in Alcohol Use Disorder: Learning from past success and failure to shape a brighter future. Neurosci Biobehav Rev 2019; 103:384-398. [PMID: 31112713 DOI: 10.1016/j.neubiorev.2019.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
Alcohol Use Disorder (AUD) is a chronic disease that develops over the years. The complexity of the neurobiological processes contributing to the emergence of AUD and the neuroadaptive changes occurring during disease progression make it difficult to improve treatments. On the other hand, this complexity offers researchers the possibility to explore new targets. Over years of intense research several molecules were tested in AUD; in most cases, despite promising preclinical data, the clinical efficacy appeared insufficient to justify futher development. A prototypical example is that of corticotropin releasing factor type 1 receptor (CRF1R) antagonists that showed significant effectiveness in animal models of AUD but were largely ineffective in humans. The present article attempts to analyze the most recent venues in the development of new medications in AUD with a focus on the most promising drug targets under current exploration. Moreover, we delineate the importance of using a more integrated translational framework approach to correlate preclinical findings and early clinical data to enhance the probability to validate biological targets of interest.
Collapse
Affiliation(s)
- Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Alessio Masi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Massimo Bramucci
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Marisa Roberto
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Angelo Bifone
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy; Department of Molecular Biotechnology and Health Science, University of Torino, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy.
| |
Collapse
|
44
|
Park CI, Hwang SS, Kim HW, Kang JI, Lee SH, Kim SJ. Association of opioid receptor gene polymorphisms with drinking severity and impulsivity related to alcohol use disorder in a Korean population. CNS Neurosci Ther 2019; 26:30-38. [PMID: 31004399 PMCID: PMC6930822 DOI: 10.1111/cns.13138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Aims Recent evidence suggests that the opioid system is implicated in the pathophysiology of alcohol use disorder (AUD). We aimed to examine the genetic influence of opioid receptors on susceptibility to AUD and its clinical and psychological characteristics including harmful drinking behavior and various aspects of impulsivity in AUD patients. Methods Three μ‐opioid receptor gene (OPRM1) variants and two κ‐opioid receptor gene (OPRK1) variants were examined in 314 male patients with AUD and 324 male controls. We applied the Alcohol Use Disorders Identification Test (AUDIT), Obsessive Compulsive Drinking Scale (OCDS), and Alcohol Dependence Scale. AUD patients also completed the stop‐signal task, delay discounting task, balloon analogue risk task, and the Barratt Impulsiveness Scale version 11 (BIS‐11). Results No significant differences in genotype distributions or haplotype frequencies were found between AUD patients and controls. However, OPRK1 SNP rs6473797 was significantly related to the severity of alcohol‐related symptoms as measured by AUDIT and OCDS and a haplotype containing rs6473797 was also related to OCDS scores in AUD patients. For other psychological traits, OPRM1 SNP rs495491 was significantly associated with scores on the motor subfactor of the BIS‐11. Conclusion Genetic variations in opioid receptors may contribute to symptom severity and impulsivity in AUD patients.
Collapse
Affiliation(s)
- Chun Il Park
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Syung Shick Hwang
- Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hae Won Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Medical Education, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee In Kang
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.,Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Joo Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Hefner KR, Starr MJ, Curtin JJ. Heavy marijuana use but not deprivation is associated with increased stressor reactivity. JOURNAL OF ABNORMAL PSYCHOLOGY 2019; 127:348-358. [PMID: 29745700 DOI: 10.1037/abn0000344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although stressors appear to motivate marijuana use, and marijuana use, in turn, is believed to induce stress system neuroadaptations, relatively little empirical work has explicitly tested for stress neuroadaptations associated with heavy marijuana use. We examined stressor reactivity to threat of unpredictable electric shock via startle potentiation among heavy marijuana users and a control group that reported minimal history of marijuana use. Heavy marijuana users were randomly assigned to 3 days of marijuana deprivation or no deprivation. This design allowed us to test contrasts for heavy (vs. minimal) use and deprivation (vs. no deprivation) on stressor reactivity. Heavy marijuana users (both deprived and nondeprived) displayed increased startle potentiation during threat of unpredictable electric shock relative to minimal use controls. In contrast, marijuana deprivation had no effect on startle potentiation. Startle potentiation was also increased among users who reported greater stress-coping motives for their marijuana use and users with cannabis use disorder diagnoses. To our knowledge, this is the 1st study to demonstrate increased reactivity to unpredictable stressors among heavy marijuana users. However, comparable increased unpredictable stressor reactivity among patients with alcohol and other substance use disorders has been previously documented. This relationship to heavy marijuana use is consistent with predictions from rodent addiction models regarding stress neuroadaptations following heavy, chronic drug use but could also represent an etiologically relevant premorbid risk characteristic. Finally, the clinical import of unpredictable stressor reactivity is reinforced by its relationships with stress-coping motives and cannabis use disorder status. (PsycINFO Database Record
Collapse
Affiliation(s)
| | - Mark J Starr
- Department of Psychology, University of Wisconsin-Madison
| | - John J Curtin
- Department of Psychology, University of Wisconsin-Madison
| |
Collapse
|
46
|
Mattalloni MS, Albrecht PA, Salinas-Luypaert C, Deza-Ponzio R, Quintanilla ME, Herrera-Marschitz M, Cancela LM, Rivera-Meza M, Virgolini MB. Silencing brain catalase expression reduces ethanol intake in developmentally-lead-exposed rats. Neurotoxicology 2019; 70:180-186. [DOI: 10.1016/j.neuro.2018.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/11/2018] [Accepted: 10/27/2018] [Indexed: 11/30/2022]
|
47
|
OPRM1 A118G and serum β-endorphin interact with sex and digit ratio (2D:4D) to influence risk and course of alcohol dependence. Eur Neuropsychopharmacol 2018; 28:1418-1428. [PMID: 30322771 DOI: 10.1016/j.euroneuro.2018.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 08/06/2018] [Accepted: 09/05/2018] [Indexed: 01/15/2023]
Abstract
Activation of mesolimbic mu-opioid receptors by their endogenous ligand, β-endorphin, can mediate the rewarding effects of alcohol. However, there is conflicting evidence on the relationship between the mu-opioid receptor (OPRM1) A118G single nucleotide polymorphism (SNP) and alcohol dependence risk. Preclinical evidence suggests that sex and sex hormone-dependent prenatal brain organization may interact with the opioid system to influence alcohol drinking behavior. We genotyped 200 alcohol-dependent patients and 240 healthy individuals for the OPRM1 A118G SNP and measured serum β-endorphin level at recruitment and after acute withdrawal. We then determined the association between these factors and alcohol dependence risk and 24-month outcome in the context of both sex and second-to-fourth digit lengths ratio (2D:4D) - a biomarker of prenatal sex hormone levels. The OPRM1 A118G AA genotype associated with elevated risk of alcohol-related hospital readmission, more readmissions, and fewer days until first readmission in male patients only. After normalizing patient 2D:4D against control 2D:4D, we found that normalized 2D:4D ratios were lower in male 118G patients than male AA patients, suggesting prenatal androgens interact with OPRM1 to influence alcohol dependence risk. In addition, β-endorphin levels after acute withdrawal correlated negatively with withdrawal severity in females but not in males, which may indicate β-endorphin protects against withdrawal-induced stress in a sex-specific manner.
Collapse
|
48
|
Jin C, Decker AM, Makhijani VH, Besheer J, Darcq E, Kieffer BL, Maitra R. Discovery of a Potent, Selective, and Brain-Penetrant Small Molecule that Activates the Orphan Receptor GPR88 and Reduces Alcohol Intake. J Med Chem 2018; 61:6748-6758. [PMID: 30011199 PMCID: PMC6108082 DOI: 10.1021/acs.jmedchem.8b00566] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The orphan G-protein-coupled receptor GPR88 is highly expressed in the striatum. Studies using GPR88 knockout mice have suggested that the receptor is implicated in alcohol seeking and drinking behaviors. To date, the biological effects of GPR88 activation are still unknown due to the lack of a potent and selective agonist appropriate for in vivo investigation. In this study, we report the discovery of the first potent, selective, and brain-penetrant GPR88 agonist RTI-13951-33 (6). RTI-13951-33 exhibited an EC50 of 25 nM in an in vitro cAMP functional assay and had no significant off-target activity at 38 GPCRs, ion channels, and neurotransmitter transporters that were tested. RTI-13951-33 displayed enhanced aqueous solubility compared to (1 R,2 R)-2-PCCA (2) and had favorable pharmacokinetic properties for behavioral assessment. Finally, RTI-13951-33 significantly reduced alcohol self-administration and alcohol intake in a dose-dependent manner without effects on locomotion and sucrose self-administration in rats when administered intraperitoneally.
Collapse
Affiliation(s)
- Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M. Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Viren H. Makhijani
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Emmanuel Darcq
- Department of Psychiatry, Douglas Mental Health Research Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Brigitte L. Kieffer
- Department of Psychiatry, Douglas Mental Health Research Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
49
|
Ben Hamida S, Mendonça-Netto S, Arefin TM, Nasseef MT, Boulos LJ, McNicholas M, Ehrlich AT, Clarke E, Moquin L, Gratton A, Darcq E, Adela HL, Maldonado R, Kieffer BL. Increased Alcohol Seeking in Mice Lacking Gpr88 Involves Dysfunctional Mesocorticolimbic Networks. Biol Psychiatry 2018; 84:202-212. [PMID: 29580570 PMCID: PMC6054571 DOI: 10.1016/j.biopsych.2018.01.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 11/28/2022]
Abstract
BACKGOUND Alcohol use disorder (AUD) is devastating and poorly treated, and innovative targets are actively sought for prevention and treatment. The orphan G protein-coupled receptor GPR88 is enriched in mesocorticolimbic pathways, and Gpr88 knockout mice show hyperactivity and risk-taking behavior, but a potential role for this receptor in drug abuse has not been examined. METHODS We tested Gpr88 knockout mice for alcohol-drinking and -seeking behaviors. To gain system-level understanding of their alcohol endophenotype, we also analyzed whole-brain functional connectivity in naïve mice using resting-state functional magnetic resonance imaging. RESULTS Gpr88 knockout mice showed increased voluntary alcohol drinking at both moderate and excessive levels, with intact alcohol sedation and metabolism. Mutant mice also showed increased operant responding and motivation for alcohol, while food and chocolate operant self-administration were unchanged. Alcohol place conditioning and alcohol-induced dopamine release in the nucleus accumbens were decreased, suggesting reduced alcohol reward in mutant mice that may partly explain enhanced alcohol drinking. Seed-based voxelwise functional connectivity analysis revealed significant remodeling of mesocorticolimbic centers, whose hallmark was predominant weakening of prefrontal cortex, ventral tegmental area, and amygdala connectional patterns. Also, effective connectivity from the ventral tegmental area to the nucleus accumbens and amygdala was reduced. CONCLUSIONS Gpr88 deletion disrupts executive, reward, and emotional networks in a configuration that reduces alcohol reward and promotes alcohol seeking and drinking. The functional connectivity signature is reminiscent of alterations observed in individuals at risk for AUD. The Gpr88 gene, therefore, may represent a vulnerability/resilience factor for AUD, and a potential drug target for AUD treatment.
Collapse
Affiliation(s)
- Sami Ben Hamida
- Département de Médecine Translationnelle et Neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U-964, CNRS UMR-7104, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France,Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Sueli Mendonça-Netto
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Tanzil Mahmud Arefin
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany,Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Md. Taufiq Nasseef
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Laura-Joy Boulos
- Département de Médecine Translationnelle et Neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U-964, CNRS UMR-7104, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France,Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Michael McNicholas
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Aliza Toby Ehrlich
- Département de Médecine Translationnelle et Neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U-964, CNRS UMR-7104, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France,Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Eleanor Clarke
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Luc Moquin
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Alain Gratton
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Emmanuel Darcq
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Harsan Laura Adela
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany,Engineering science, computer science and imaging laboratory (ICube), Integrative Multimodal Imaging in Healthcare, University of Strasbourg – CNRS, Strasbourg, France,Department of Biophysics and Nuclear Medicine, Faculty of Medicine, University Hospital Strasbourg, Strasbourg, France
| | - Rafael Maldonado
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Brigitte Lina Kieffer
- Département de Médecine Translationnelle et Neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U-964, Centre National de la Recherche Scientifique UMR-7104, University of Strasbourg, Illkirch-Graffenstaden, Strasbourg, France; Douglas Mental Health Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
50
|
Bazov I, Sarkisyan D, Kononenko O, Watanabe H, Karpyak VM, Yakovleva T, Bakalkin G. Downregulation of the neuronal opioid gene expression concomitantly with neuronal decline in dorsolateral prefrontal cortex of human alcoholics. Transl Psychiatry 2018; 8:122. [PMID: 29925858 PMCID: PMC6010434 DOI: 10.1038/s41398-017-0075-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/02/2017] [Accepted: 11/01/2017] [Indexed: 01/03/2023] Open
Abstract
Molecular changes in cortical areas of addicted brain may underlie cognitive impairment and loss of control over intake of addictive substances and alcohol. Prodynorphin (PDYN) gives rise to dynorphin (DYNs) opioid peptides which target kappa-opioid receptor (KOR). DYNs mediate alcohol-induced impairment of learning and memory, while KOR antagonists block excessive, compulsive-like drug and alcohol self-administration in animal models. In human brain, the DYN/KOR system may undergo adaptive changes, which along with neuronal loss, may contribute to alcohol-associated cognitive deficit. We addressed this hypothesis by comparing the expression levels and co-expression (transcriptionally coordinated) patterns of PDYN and KOR (OPRK1) genes in dorsolateral prefrontal cortex (dlPFC) between human alcoholics and controls. Postmortem brain specimens of 53 alcoholics and 55 controls were analyzed. PDYN was found to be downregulated in dlPFC of alcoholics, while OPRK1 transcription was not altered. PDYN downregulation was confined to subgroup of subjects carrying C, a high-risk allele of PDYN promoter SNP rs1997794 associated with alcoholism. Changes in PDYN expression did not depend on the decline in neuronal proportion in alcoholics, and thereby may be attributed to transcriptional adaptations in alcoholic brain. Absolute expression levels of PDYN were lower compared to those of OPRK1, suggesting that PDYN expression is a limiting factor in the DYN/KOR signaling, and that the PDYN downregulation diminishes efficacy of DYN/KOR signaling in dlPFC of human alcoholics. The overall outcome of the DYN/KOR downregulation may be disinhibition of neurotransmission, which when overactivated could contribute to formation of alcohol-related behavior.
Collapse
Affiliation(s)
- Igor Bazov
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24, Uppsala, Sweden.
| | - Daniil Sarkisyan
- 0000 0004 1936 9457grid.8993.bDivision of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Olga Kononenko
- 0000 0004 1936 9457grid.8993.bDivision of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Hiroyuki Watanabe
- 0000 0004 1936 9457grid.8993.bDivision of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Victor M. Karpyak
- 0000 0004 0459 167Xgrid.66875.3aDepartment of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Tatiana Yakovleva
- 0000 0004 1936 9457grid.8993.bDivision of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Georgy Bakalkin
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 24, Uppsala, Sweden.
| |
Collapse
|