1
|
Shi X, Wang Y, Yang Z, Yuan W, Li MD. Identification and validation of a novel gene ARVCF associated with alcohol dependence among Chinese population. iScience 2024; 27:110976. [PMID: 39429782 PMCID: PMC11490727 DOI: 10.1016/j.isci.2024.110976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Alcohol dependence is a heritable disorder, yet its genetic basis and underlying mechanisms remain poorly understood, especially in Chinese population. In this study, we conducted gene-based and transcript-based association tests and found a significant association between ARVCF expression in the cortex and hippocampus of the brain and alcohol use in a cohort of 1,329 individuals with Chinese ancestry. Further analysis using the effective-median-based Mendelian randomization framework for inferring the causal genes (EMIC) revealed a causal relationship between ARVCF expression in the frontal cortex and alcohol use. Moreover, leveraging extensive European alcohol dependence data, our gene association tests and EMIC analysis showed that ARVCF expression in the nucleus accumbens was significantly associated with alcohol dependence. Finally, animal studies indicated that Arvcf knockout mice lacked conditioned place preference for alcohol. Together, our combined human genetic and animal studies indicate that ARVCF plays a crucial role in alcohol dependence.
Collapse
Affiliation(s)
- Xiaoqiang Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wenji Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ming D. Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Research Center for Air Pollution and Health, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Trang KB, Chesi A, Toikumo S, Pippin JA, Pahl MC, O’Brien JM, Amundadottir LT, Brown KM, Yang W, Welles J, Santoleri D, Titchenell PM, Seale P, Zemel BS, Wagley Y, Hankenson KD, Kaestner KH, Anderson SA, Kayser MS, Wells AD, Kranzler HR, Kember RL, Grant SF. Shared and unique 3D genomic features of substance use disorders across multiple cell types. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.18.24310649. [PMID: 39072016 PMCID: PMC11275669 DOI: 10.1101/2024.07.18.24310649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Recent genome-wide association studies (GWAS) have revealed shared genetic components among alcohol, opioid, tobacco and cannabis use disorders. However, the extent of the underlying shared causal variants and effector genes, along with their cellular context, remain unclear. We leveraged our existing 3D genomic datasets comprising high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq and RNA-seq across >50 diverse human cell types to focus on genomic regions that coincide with GWAS loci. Using stratified LD regression, we determined the proportion of genomewide SNP heritability attributable to the features assayed across our cell types by integrating recent GWAS summary statistics for the relevant traits: alcohol use disorder (AUD), tobacco use disorder (TUD), opioid use disorder (OUD) and cannabis use disorder (CanUD). Statistically significant enrichments (P<0.05) were observed in 14 specific cell types, with heritability reaching 9.2-fold for iPSC-derived cortical neurons and neural progenitors, confirming that they are crucial cell types for further functional exploration. Additionally, several pancreatic cell types, notably pancreatic beta cells, showed enrichment for TUD, with heritability enrichments up to 4.8-fold, suggesting genomic overlap with metabolic processes. Further investigation revealed significant positive genetic correlations between T2D with both TUD and CanUD (FDR<0.05) and a significant negative genetic correlation with AUD. Interestingly, after partitioning the heritability for each cell type's cis-regulatory elements, the correlation between T2D and TUD for pancreatic beta cells was greater (r=0.2) than the global genetic correlation value. Our study provides new genomic insights into substance use disorders and implicates cell types where functional follow-up studies could reveal causal variant-gene mechanisms underpinning these disorders.
Collapse
Affiliation(s)
- Khanh B. Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joan M. O’Brien
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Disease, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M. Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wenli Yang
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaclyn Welles
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominic Santoleri
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yadav Wagley
- Department of Orthopedic Surgery, University of Michigan Medical School Ann Arbor, MI, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School Ann Arbor, MI, USA
| | - Klaus H. Kaestner
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew S. Kayser
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel L. Kember
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
3
|
Tarantino G, Citro V. What are the common downstream molecular events between alcoholic and nonalcoholic fatty liver? Lipids Health Dis 2024; 23:41. [PMID: 38331795 PMCID: PMC10851522 DOI: 10.1186/s12944-024-02031-1] [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: 11/05/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Liver fat storage, also called hepatic steatosis, is increasingly common and represents a very frequent diagnosis in the medical field. Excess fat is not without consequences. In fact, hepatic steatosis contributes to the progression toward liver fibrosis. There are two main types of fatty liver disease, alcoholic fatty liver disease (AFLD) and nonalcoholic fatty liver disease (NAFLD). Although AFLD and NAFLD are similar in their initial morphological features, both conditions involve the same evolutive forms. Moreover, there are various common mechanisms underlying both diseases, including alcoholic liver disease and NAFLD, which are commonalities. In this Review, the authors explore similar downstream signaling events involved in the onset and progression of the two entities but not completely different entities, predominantly focusing on the gut microbiome. Downstream molecular events, such as the roles of sirtuins, cytokeratins, adipokines and others, should be considered. Finally, to complete the feature, some new tendencies in the therapeutic approach are presented.
Collapse
Affiliation(s)
| | - Vincenzo Citro
- Department of General Medicine, Umberto I Hospital, Nocera Inferiore, SA, 84014, Italy
| |
Collapse
|
4
|
Kember RL, Hartwell EE, Xu H, Rotenberg J, Almasy L, Zhou H, Gelernter J, Kranzler HR. Phenome-wide Association Analysis of Substance Use Disorders in a Deeply Phenotyped Sample. Biol Psychiatry 2023; 93:536-545. [PMID: 36273948 PMCID: PMC9931661 DOI: 10.1016/j.biopsych.2022.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/17/2022] [Accepted: 08/05/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Substance use disorders (SUDs) are associated with a variety of co-occurring psychiatric disorders and other SUDs, which partly reflects genetic pleiotropy. Polygenic risk scores (PRSs) and phenome-wide association studies are useful in evaluating pleiotropic effects. However, the comparatively low prevalence of SUDs in population samples and the lack of detailed information available in electronic health records limit these data sets' informativeness for such analyses. METHODS We used the deeply phenotyped Yale-Penn sample (n = 10,610 with genetic data; 46.3% African ancestry, 53.7% European ancestry) to examine pleiotropy for 4 major substance-related traits: alcohol use disorder, opioid use disorder, smoking initiation, and lifetime cannabis use. The sample includes both affected and control subjects interviewed using the Semi-Structured Assessment for Drug Dependence and Alcoholism, a comprehensive psychiatric interview. RESULTS In African ancestry individuals, PRS for alcohol use disorder, and in European individuals, PRS for alcohol use disorder, opioid use disorder, and smoking initiation were associated with their respective primary DSM diagnoses. These PRSs were also associated with additional phenotypes involving the same substance. Phenome-wide association study analyses of PRS in European individuals identified associations across multiple phenotypic domains, including phenotypes not commonly assessed in phenome-wide association study analyses, such as family environment and early childhood experiences. CONCLUSIONS Smaller, deeply phenotyped samples can complement large biobank genetic studies with limited phenotyping by providing greater phenotypic granularity. These efforts allow associations to be identified between specific features of disorders and genetic liability for SUDs, which help to inform our understanding of the pleiotropic pathways underlying them.
Collapse
Affiliation(s)
- Rachel L Kember
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Mental Illness Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania.
| | - Emily E Hartwell
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Mental Illness Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Heng Xu
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - James Rotenberg
- Mental Illness Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; VA Connecticut Healthcare System, West Haven, Connecticut
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; VA Connecticut Healthcare System, West Haven, Connecticut; Departments of Genetics and Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Mental Illness Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Sabotta CM, Kwan SY, Petty LE, Below JE, Joon A, Wei P, Fisher-Hoch SP, McCormick JB, Beretta L. Genetic variants associated with circulating liver injury markers in Mexican Americans, a population at risk for non-alcoholic fatty liver disease. Front Genet 2022; 13:995488. [PMID: 36386790 PMCID: PMC9644071 DOI: 10.3389/fgene.2022.995488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/10/2022] [Indexed: 02/03/2023] Open
Abstract
Objective: Mexican Americans are disproportionally affected by non-alcoholic fatty liver disease (NAFLD), liver fibrosis and hepatocellular carcinoma. Noninvasive means to identify those in this population at high risk for these diseases are urgently needed. Approach: The Cameron County Hispanic Cohort (CCHC) is a population-based cohort with high rates of obesity (51%), type 2 diabetes (28%) and NAFLD (49%). In a subgroup of 564 CCHC subjects, we evaluated 339 genetic variants previously reported to be associated with liver injury markers aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in United Kingdom and Japanese cohorts. Results: Association was confirmed for 86 variants. Among them, 27 had higher effect allele frequency in the CCHC than in the United Kingdom and Japanese cohorts, and 16 had stronger associations with AST and ALT than rs738409 (PNPLA3). These included rs17710008 (MYCT1), rs2519093 (ABO), rs1801690 (APOH), rs10409243 (S1PR2), rs1800759 (LOC100507053) and rs2491441 (RGL1), which were also associated with steatosis and/or liver fibrosis measured by vibration-controlled transient elastography. Main contributors to advanced fibrosis risk were rs11240351 (CNTN2), rs1800759 (LOC100507053), rs738409 (PNPLA3) and rs1801690 (APOH), with advanced fibrosis detected in 37.5% of subjects with 3 of these 4 variants [AOR = 11.6 (95% CI) = 3.8-35.3]. AST- and ALT-associated variants implicated distinct pathways (ethanol and galactose degradation versus antigen presentation and B cell development). Finally, 8 variants, including rs62292950 (DNAJC13), were associated with gut microbiome changes. Conclusion: These genotype-phenotype findings may have utility in risk modeling and disease prevention in this high-risk population.
Collapse
Affiliation(s)
- Caroline M. Sabotta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Suet-Ying Kwan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lauren E. Petty
- Vanderbilt Genetics Institute and Department of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jennifer E. Below
- Vanderbilt Genetics Institute and Department of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Aron Joon
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Susan P. Fisher-Hoch
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, United States
| | - Joseph B. McCormick
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, United States
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
6
|
Denham AN, Drake J, Gavrilov M, Taylor ZN, Bacanu SA, Vladimirov VI. Long Non-Coding RNAs: The New Frontier into Understanding the Etiology of Alcohol Use Disorder. Noncoding RNA 2022; 8:59. [PMID: 36005827 PMCID: PMC9415279 DOI: 10.3390/ncrna8040059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022] Open
Abstract
Alcohol use disorder (AUD) is a complex, chronic, debilitating condition impacting millions worldwide. Genetic, environmental, and epigenetic factors are known to contribute to the development of AUD. Long non-coding RNAs (lncRNAs) are a class of regulatory RNAs, commonly referred to as the "dark matter" of the genome, with little to no protein-coding potential. LncRNAs have been implicated in numerous processes critical for cell survival, suggesting that they play important functional roles in regulating different cell processes. LncRNAs were also shown to display higher tissue specificity than protein-coding genes and have a higher abundance in the brain and central nervous system, demonstrating a possible role in the etiology of psychiatric disorders. Indeed, genetic (e.g., genome-wide association studies (GWAS)), molecular (e.g., expression quantitative trait loci (eQTL)) and epigenetic studies from postmortem brain tissues have identified a growing list of lncRNAs associated with neuropsychiatric and substance use disorders. Given that the expression patterns of lncRNAs have been associated with widespread changes in the transcriptome, including methylation, chromatin architecture, and activation or suppression of translational activity, the regulatory nature of lncRNAs may be ubiquitous and an innate component of gene regulation. In this review, we present a synopsis of the functional impact that lncRNAs may play in the etiology of AUD. We also discuss the classifications of lncRNAs, their known functional roles, and therapeutic advancements in the field of lncRNAs to further clarify the functional relationship between lncRNAs and AUD.
Collapse
Affiliation(s)
- Allie N. Denham
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX 77807, USA
- Department of Psychiatry, College of Medicine, University of Arizona Phoenix, Phoenix, AZ 85004, USA
| | - John Drake
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX 77807, USA
- Department of Psychiatry, College of Medicine, University of Arizona Phoenix, Phoenix, AZ 85004, USA
- MSCI Program, Texas A&M University, Bryan, TX 77807, USA
| | - Matthew Gavrilov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX 77807, USA
| | - Zachary N. Taylor
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX 77807, USA
- Department of Psychiatry, College of Medicine, University of Arizona Phoenix, Phoenix, AZ 85004, USA
| | - Silviu-Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23219, USA
- Departent of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Vladimir I. Vladimirov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX 77807, USA
- Department of Psychiatry, College of Medicine, University of Arizona Phoenix, Phoenix, AZ 85004, USA
- Departent of Psychiatry, Virginia Commonwealth University, Richmond, VA 23219, USA
- Texas A&M Institute for Neuroscience, College Station, Texas A&M University, College Station, TX 77843, USA
- Genetics Interdisciplinary Program, College Station, Texas A&M University, College Station, TX 77843, USA
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
de Marco A, Scozia G, Manfredi L, Conversi D. A Systematic Review of Genetic Polymorphisms Associated with Bipolar Disorder Comorbid to Substance Abuse. Genes (Basel) 2022; 13:genes13081303. [PMID: 35893041 PMCID: PMC9330731 DOI: 10.3390/genes13081303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 01/09/2023] Open
Abstract
It is currently unknown which genetic polymorphisms are involved in substance use disorder (SUD) comorbid with bipolar disorder (BD). The research on polymorphisms in BD comorbid with SUD (BD + SUD) is summarized in this systematic review. We looked for case-control studies that genetically compared adults and adolescents with BD and SUD, healthy controls, and BD without SUD. PRISMA was used to create our protocol, which is PROSPERO-registered (identification: CRD4221270818). The following bibliographic databases were searched indefinitely until December 2021 to identify potentially relevant articles: PubMed, PsycINFO, Scopus, and Web of Science. This systematic review, after the qualitative analysis of the study selection, included 17 eligible articles. In the selected studies, 66 polymorphisms in 29 genes were investigated. The present work delivers a group of potentially valuable genetic polymorphisms associated with BD + SUD: rs11600996 (ARNTL), rs228642/rs228682/rs2640909 (PER3), PONQ192R (PON1), rs945032 (BDKRB2), rs1131339 (NR4A3), and rs6971 (TSPO). It is important to note that none of those findings have been confirmed by two or more studies; thus, we believe that all the polymorphisms identified in this review require additional evidence to be confirmed.
Collapse
Affiliation(s)
- Adriano de Marco
- Department of Psychology, Università degli Studi di Roma ‘La Sapienza’, 00185 Rome, Italy; (A.d.M.); (G.S.); (L.M.)
| | - Gabriele Scozia
- Department of Psychology, Università degli Studi di Roma ‘La Sapienza’, 00185 Rome, Italy; (A.d.M.); (G.S.); (L.M.)
- PhD Program in Behavioral Neuroscience, Università degli Studi di Roma ‘La Sapienza’, 00185 Rome, Italy
| | - Lucia Manfredi
- Department of Psychology, Università degli Studi di Roma ‘La Sapienza’, 00185 Rome, Italy; (A.d.M.); (G.S.); (L.M.)
| | - David Conversi
- Department of Psychology, Università degli Studi di Roma ‘La Sapienza’, 00185 Rome, Italy; (A.d.M.); (G.S.); (L.M.)
- Correspondence:
| |
Collapse
|
8
|
Alcohol-Induced Oxidative Stress and the Role of Antioxidants in Alcohol Use Disorder: A Systematic Review. Antioxidants (Basel) 2022; 11:antiox11071374. [PMID: 35883865 PMCID: PMC9311529 DOI: 10.3390/antiox11071374] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a highly prevalent, comorbid, and disabling disorder. The underlying mechanism of ethanol neurotoxicity and the involvement of oxidative stress is still not fully elucidated. However, ethanol metabolism has been associated with increased oxidative stress through alcohol dehydrogenase, the microsomal ethanol oxidation system, and catalase metabolic pathways. We searched the PubMed and genome-wide association studies (GWAS) catalog databases to review the literature systematically and summarized the findings focusing on AUD and alcohol abstinence in relation to oxidative stress. In addition, we reviewed the ClinicalTrials.gov resource of the US National Library of Medicine to identify all ongoing and completed clinical trials that include therapeutic interventions based on antioxidants. The retrieved clinical and preclinical studies show that oxidative stress impacts AUD through genetics, alcohol metabolism, inflammation, and neurodegeneration.
Collapse
|
9
|
Schaschl H, Göllner T, Morris DL. Positive selection acts on regulatory genetic variants in populations of European ancestry that affect ALDH2 gene expression. Sci Rep 2022; 12:4563. [PMID: 35296751 PMCID: PMC8927298 DOI: 10.1038/s41598-022-08588-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/09/2022] [Indexed: 11/09/2022] Open
Abstract
ALDH2 is a key enzyme in alcohol metabolism that protects cells from acetaldehyde toxicity. Using iHS, iSAFE and FST statistics, we identified regulatory acting variants affecting ALDH2 gene expression under positive selection in populations of European ancestry. Several SNPs (rs3184504, rs4766578, rs10774625, rs597808, rs653178, rs847892, rs2013002) that function as eQTLs for ALDH2 in various tissues showed evidence of strong positive selection. Very large pairwise FST values indicated high genetic differentiation at these loci between populations of European ancestry and populations of other global ancestries. Estimating the timing of positive selection on the beneficial alleles suggests that these variants were recently adapted approximately 3000-3700 years ago. The derived beneficial alleles are in complete linkage disequilibrium with the derived ALDH2 promoter variant rs886205, which is associated with higher transcriptional activity. The SNPs rs4766578 and rs847892 are located in binding sequences for the transcription factor HNF4A, which is an important regulatory element of ALDH2 gene expression. In contrast to the missense variant ALDH2 rs671 (ALDH2*2), which is common only in East Asian populations and is associated with greatly reduced enzyme activity and alcohol intolerance, the beneficial alleles of the regulatory variants identified in this study are associated with increased expression of ALDH2. This suggests adaptation of Europeans to higher alcohol consumption.
Collapse
Affiliation(s)
- Helmut Schaschl
- Department of Evolutionary Anthropology, Faculty of Life Sciences, University of Vienna, Djerassiplatz 1, 1030, Vienna, Austria.
| | - Tobias Göllner
- Department of Evolutionary Anthropology, Faculty of Life Sciences, University of Vienna, Djerassiplatz 1, 1030, Vienna, Austria
| | - David L Morris
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, Great Maze Pond, London, SE1 9RT, UK
| |
Collapse
|
10
|
Plawecki MH, Boes J, Wetherill L, Kosobud AEK, Stangl BL, Ramchandani VA, Zimmermann US, Nurnberger JI, Schuckit M, Edenberg HJ, Pandey G, Kamarajan C, Porjesz B, Foroud T, O'Connor S. Binge and high-intensity drinking-Associations with intravenous alcohol self-administration and underlying risk factors. Addict Biol 2022; 27:e13228. [PMID: 36301209 PMCID: PMC9786574 DOI: 10.1111/adb.13228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/06/2022] [Accepted: 08/14/2022] [Indexed: 01/24/2023]
Abstract
Some styles of alcohol consumption are riskier than others. How the level and rate of alcohol exposure contribute to the increased risk of alcohol use disorder is unclear, but likely depends on the alcohol concentration time course. We hypothesized that the brain is sensitive to the alcohol concentration rate of change and that people at greater risk would self-administer faster. We developed a novel intravenous alcohol self-administration paradigm to allow participants direct and reproducible control over how quickly their breath alcohol concentration changes. We used drinking intensity and the density of biological family history of alcohol dependence as proxies for risk. Thirty-five alcohol drinking participants aged 21-28 years provided analytical data from a single, intravenous alcohol self-administration session using our computer-assisted alcohol infusion system rate control paradigm. A shorter time to reach 80 mg/dl was associated with increasing multiples of the binge drinking definition (p = 0.004), which was in turn related to higher density of family history of alcoholism (FHD, p = 0.04). Rate-dependent changes in subjective response (intoxication and stimulation) were also associated with FHD (each p = 0.001). Subsequently, given the limited sample size and FHD range, associations between multiples of the binge drinking definition and FHD were replicated and extended in analyses of the Collaborative Study on the Genetics of Alcoholism database. The rate control paradigm models binge and high-intensity drinking in the laboratory and provides a novel way to examine the relationship between the pharmacokinetics and pharmacodynamics of alcohol and potentially the risk for the development of alcohol use disorders.
Collapse
Affiliation(s)
- Martin H. Plawecki
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Julian Boes
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leah Wetherill
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ann E. K. Kosobud
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Bethany L. Stangl
- Human Psychopharmacology Laboratory, Division of Intramural Clinical and Biological ResearchNIAAABethesdaMarylandUSA
| | - Vijay A. Ramchandani
- Human Psychopharmacology Laboratory, Division of Intramural Clinical and Biological ResearchNIAAABethesdaMarylandUSA
| | - Ulrich S. Zimmermann
- Department of Psychiatry and PsychotherapyUniversity Hospital Carl Gustav Carus of the Technische Universität DresdenDresdenGermany,Department of Addiction Medicine and Psychotherapykbo Isar‐Amper‐Klinikum Haar/MunichMunichGermany
| | - John I. Nurnberger
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Marc Schuckit
- Department of PsychiatryUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Howard J. Edenberg
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA,Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gayathri Pandey
- Henri Begleiter Neurodynamics LaboratoryState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Chella Kamarajan
- Henri Begleiter Neurodynamics LaboratoryState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Bernice Porjesz
- Henri Begleiter Neurodynamics LaboratoryState University of New York Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sean O'Connor
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
11
|
Yan Y, Wang J, Yu L, Cui B, Wang H, Xiao X, Zhang Y, Zheng J, Wang J, Hui R, Wang Y. ANKRD36 Is Involved in Hypertension by Altering Expression of ENaC Genes. Circ Res 2021; 129:1067-1081. [PMID: 34615377 DOI: 10.1161/circresaha.121.319883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yupeng Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Jin'e Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Liang Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Hongrui Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Xiao Xiao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Jun Zheng
- Rizhao Port Hospital, Shandong, China (J.Z., Jingjun Wang)
| | - Jingjun Wang
- Rizhao Port Hospital, Shandong, China (J.Z., Jingjun Wang)
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| |
Collapse
|
12
|
Rodriguez FD. Targeting Epigenetic Mechanisms to Treat Alcohol Use Disorders (AUD). Curr Pharm Des 2021; 27:3252-3272. [PMID: 33535943 PMCID: PMC8778698 DOI: 10.2174/1381612827666210203142539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/08/2020] [Indexed: 12/04/2022]
Abstract
BACKGROUND The impact of abusive alcohol consumption on human health is remarkable. According to the World Health Organization (WHO), approximately 3.3 million people die annually because of harmful alcohol consumption (the figure represents around 5.9% of global deaths). Alcohol Use Disorder (AUD) is a chronic disease where individuals exhibit compulsive alcohol drinking and present negative emotional states when they do not drink. In the most severe manifestations of AUD, the individuals lose control over intake despite a decided will to stop drinking. Given the multiple faces and the specific forms of this disease, the term AUD often appears in the plural (AUDs). Since only a few approved pharmacological treatments are available to treat AUD and they do not apply to all individuals or AUD forms, the search for compounds that may help to eliminate the burden of the disease and complement other therapeutical approaches is necessary. METHODS This work reviews recent research focused on the involvement of epigenetic mechanisms in the pathophysiology of AUD. Excessive drinking leads to chronic and compulsive consumption that eventually damages the organism. The central nervous system is a key target and is the focus of this study. The search for the genetic and epigenetic mechanisms behind the intricated dysregulation induced by ethanol will aid researchers in establishing new therapy approaches. CONCLUSION Recent findings in the field of epigenetics are essential and offer new windows for observation and research. The study of small molecules that inhibit key epienzymes involved in nucleosome architecture dynamics is necessary in order to prove their action and specificity in the laboratory and to test their effectivity and safety in clinical trials with selected patients bearing defined alterations caused by ethanol.
Collapse
Affiliation(s)
- F. David Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, University of Salamanca and Group GIR BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
| |
Collapse
|
13
|
Abstract
Substance use disorders (SUDs) are prevalent and result in an array of negative consequences. They are influenced by genetic factors (h2 = ~50%). Recent years have brought substantial progress in our understanding of the genetic etiology of SUDs and related traits. The present review covers the current state of the field for SUD genetics, including the epidemiology and genetic epidemiology of SUDs, findings from the first-generation of SUD genome-wide association studies (GWAS), cautions about translating GWAS findings to clinical settings, and suggested prioritizations for the next wave of SUD genetics efforts. Recent advances in SUD genetics have been facilitated by the assembly of large GWAS samples, and the development of state-of-the-art methods modeling the aggregate effect of genome-wide variation. These advances have confirmed that SUDs are highly polygenic with many variants across the genome conferring risk, the vast majority of which are of small effect. Downstream analyses have enabled finer resolution of the genetic architecture of SUDs and revealed insights into their genetic relationship with other psychiatric disorders. Recent efforts have also prioritized a closer examination of GWAS findings that have suggested non-uniform genetic influences across measures of substance use (e.g. consumption) and problematic use (e.g. SUD). Additional highlights from recent SUD GWAS include the robust confirmation of loci in alcohol metabolizing genes (e.g. ADH1B and ALDH2) affecting alcohol-related traits, and loci within the CHRNA5-CHRNA3-CHRNB4 gene cluster influencing nicotine-related traits. Similar successes are expected for cannabis, opioid, and cocaine use disorders as sample sizes approach those assembled for alcohol and nicotine.
Collapse
Affiliation(s)
- Joseph D. Deak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Emma C. Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Clergue-Duval V, Nicolas-Sacy L, Karsinti E, Zerdazi EH, Laplanche JL, Brousse G, Marees AT, Derks EM, Henry P, Bellivier F, Vorspan F, Bloch V. Risk and Protective Factors of Lifetime Cocaine-Associated Chest Pain. Front Psychiatry 2021; 12:704276. [PMID: 34366936 PMCID: PMC8335401 DOI: 10.3389/fpsyt.2021.704276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Cocaine users often present with repetitive events of cocaine-associated chest pain (CACP), clinically resembling acute coronary syndromes. The aim of the study is to describe the specific risk factors for CACP. Method: Cocaine users (n = 316) were recruited for a multicenter cross-sectional study. Lifetime CACP history, sociodemographic factors, and lifetime use of cocaine and other substances were assessed. Thirty single nucleotide polymorphisms (SNPs) of NOS3, ROCK2, EDN1, GUCY1A3, and ALDH2 genes, suggested by the literature on coronary spasms, were selected. The associations with CACP history were tested using the chi-square test, Student's t-test and logistic regression. Results: Among the 316 subjects [78.5% men, mean age 37.5 years, (standard-deviation ±8.7)], 190 (60.1%) were daily cocaine users and 103 (32.6%) reported a lifetime CACP history. Among those with a lifetime CACP history, the median was 10 events per individual. In multivariate analysis, lifetime CACP history was associated with daily cocaine use [odds-ratio (OR) 3.24; 95% confidence intervals (1.29-9.33)], rapid route of cocaine use [OR 2.33 (1.20-4.64) vs. intranasal use], and lifetime amphetamine use [daily amphetamine use: OR 2.80 (1.25-6.32) and non-daily amphetamine use: OR 2.14 (1.15-4.04) vs. never used]. Patients with lifetime opioid maintenance treatment (OMT) reported significantly less lifetime CACP history [OR 0.35 (0.16-0.76)]. None of the selected SNPs was associated with CACP history after multiple testing corrections. Conclusions: Clinical variables describing the intensity of stimulant use were positively associated with lifetime CACP history, while OMT was negatively associated with it. Specific harm reduction strategies can target these risk factors.
Collapse
Affiliation(s)
- Virgile Clergue-Duval
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Louise Nicolas-Sacy
- APHP, Pharmacie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
| | - Emily Karsinti
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Laboratoire ClipsyD, Université Paris Nanterre, Nanterre, France
| | - El-Hadi Zerdazi
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- APHP, Service d'addictologie, DMU IMPACT, GHU Mondor, Hôpital Emile ROUX, Limeil Brévannes, France
| | - Jean-Louis Laplanche
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- APHP, Département de Biochimie et Biologie Moléculaire, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
- Faculté de Pharmacie, Université de Paris, Paris, France
| | - Georges Brousse
- Service de psychiatrie-addictologie, CHU de Clermont-Ferrand, Université Clermont-Auvergne, Clermont-Ferrand, France
| | - Andries T. Marees
- Department of Economics, School of Business and Economics, VU University Amsterdam, Amsterdam, Netherlands
| | - Eske M. Derks
- Queensland Institute of Medical Research Berghofer, Translational Neurogenomics Group, Brisbane, QLD, Australia
| | - Patrick Henry
- Faculté de Médecine, Université de Paris, Paris, France
- APHP, Département de Cardiologie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
| | - Frank Bellivier
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Florence Vorspan
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Vanessa Bloch
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- APHP, Pharmacie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
- Faculté de Pharmacie, Université de Paris, Paris, France
| |
Collapse
|
15
|
Converging vulnerability factors for compulsive food and drug use. Neuropharmacology 2021; 196:108556. [PMID: 33862029 DOI: 10.1016/j.neuropharm.2021.108556] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022]
Abstract
Highly palatable foods and substance of abuse have intersecting neurobiological, metabolic and behavioral effects relevant for understanding vulnerability to conditions related to food (e.g., obesity, binge eating disorder) and drug (e.g., substance use disorder) misuse. Here, we review data from animal models, clinical populations and epidemiological evidence in behavioral, genetic, pathophysiologic and therapeutic domains. Results suggest that consumption of highly palatable food and drugs of abuse both impact and conversely are regulated by metabolic hormones and metabolic status. Palatable foods high in fat and/or sugar can elicit adaptation in brain reward and withdrawal circuitry akin to substances of abuse. Intake of or withdrawal from palatable food can impact behavioral sensitivity to drugs of abuse and vice versa. A robust literature suggests common substrates and roles for negative reinforcement, negative affect, negative urgency, and impulse control deficits, with both highly palatable foods and substances of abuse. Candidate genetic risk loci shared by obesity and alcohol use disorders have been identified in molecules classically associated with both metabolic and motivational functions. Finally, certain drugs may have overlapping therapeutic potential to treat obesity, diabetes, binge-related eating disorders and substance use disorders. Taken together, data are consistent with the hypotheses that compulsive food and substance use share overlapping, interacting substrates at neurobiological and metabolic levels and that motivated behavior associated with feeding or substance use might constitute vulnerability factors for one another. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
|
16
|
Kinreich S, Meyers JL, Maron-Katz A, Kamarajan C, Pandey AK, Chorlian DB, Zhang J, Pandey G, Subbie-Saenz de Viteri S, Pitti D, Anokhin AP, Bauer L, Hesselbrock V, Schuckit MA, Edenberg HJ, Porjesz B. Predicting risk for Alcohol Use Disorder using longitudinal data with multimodal biomarkers and family history: a machine learning study. Mol Psychiatry 2021; 26:1133-1141. [PMID: 31595034 PMCID: PMC7138692 DOI: 10.1038/s41380-019-0534-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 11/09/2022]
Abstract
Predictive models have succeeded in distinguishing between individuals with Alcohol use Disorder (AUD) and controls. However, predictive models identifying who is prone to develop AUD and the biomarkers indicating a predisposition to AUD are still unclear. Our sample (n = 656) included offspring and non-offspring of European American (EA) and African American (AA) ancestry from the Collaborative Study of the Genetics of Alcoholism (COGA) who were recruited as early as age 12 and were unaffected at first assessment and reassessed years later as AUD (DSM-5) (n = 328) or unaffected (n = 328). Machine learning analysis was performed for 220 EEG measures, 149 alcohol-related single nucleotide polymorphisms (SNPs) from a recent large Genome-wide Association Study (GWAS) of alcohol use/misuse and two family history (mother DSM-5 AUD and father DSM-5 AUD) features using supervised, Linear Support Vector Machine (SVM) classifier to test which features assessed before developing AUD predict those who go on to develop AUD. Age, gender, and ancestry stratified analyses were performed. Results indicate significant and higher accuracy rates for the AA compared with the EA prediction models and a higher model accuracy trend among females compared with males for both ancestries. Combined EEG and SNP features model outperformed models based on only EEG features or only SNP features for both EA and AA samples. This multidimensional superiority was confirmed in a follow-up analysis in the AA age groups (12-15, 16-19, 20-30) and EA age group (16-19). In both ancestry samples, the youngest age group achieved higher accuracy score than the two other older age groups. Maternal AUD increased the model's accuracy in both ancestries' samples. Several discriminative EEG measures and SNPs features were identified, including lower posterior gamma, higher slow wave connectivity (delta, theta, alpha), higher frontal gamma ratio, higher beta correlation in the parietal area, and 5 SNPs: rs4780836, rs2605140, rs11690265, rs692854, and rs13380649. Results highlight the significance of sampling uniformity followed by stratified (e.g., ancestry, gender, developmental period) analysis, and wider selection of features, to generate better prediction scores allowing a more accurate estimation of AUD development.
Collapse
Affiliation(s)
- Sivan Kinreich
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA.
| | - Jacquelyn L Meyers
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Adi Maron-Katz
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Chella Kamarajan
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Ashwini K Pandey
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - David B Chorlian
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Jian Zhang
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Gayathri Pandey
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | | | - Dan Pitti
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Andrey P Anokhin
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Lance Bauer
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Victor Hesselbrock
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Marc A Schuckit
- Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bernice Porjesz
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
17
|
Choquet H, Yin J, Jorgenson E. Cigarette smoking behaviors and the importance of ethnicity and genetic ancestry. Transl Psychiatry 2021; 11:120. [PMID: 33633108 PMCID: PMC7907280 DOI: 10.1038/s41398-021-01244-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 11/09/2022] Open
Abstract
Cigarette smoking contributes to numerous diseases and is one of the leading causes of death in the United States. Smoking behaviors vary widely across race/ethnicity, but it is not clear why. Here, we examine the contribution of genetic ancestry to variation in two smoking-related traits in 43,485 individuals from four race/ethnicity groups (non-Hispanic white, Hispanic/Latino, East Asian, and African American) from a single U.S. healthcare plan. Smoking prevalence was the lowest among East Asians (22.7%) and the highest among non-Hispanic whites (38.5%). We observed significant associations between genetic ancestry and smoking-related traits. Within East Asians, we observed higher smoking prevalence with greater European (versus Asian) ancestry (P = 9.95 × 10-12). Within Hispanic/Latinos, higher cigarettes per day (CPD) was associated with greater European ancestry (P = 3.34 × 10-25). Within non-Hispanic whites, the lowest number of CPD was observed for individuals of southeastern European ancestry (P = 9.06 × 10-5). These associations remained after considering known smoking-associated loci, education, socioeconomic factors, and marital status. Our findings support the role of genetic ancestry and socioeconomic factors in cigarette smoking behaviors in non-Hispanic whites, Hispanic/Latinos, and East Asians.
Collapse
Affiliation(s)
- Hélène Choquet
- Division of Research, Kaiser Permanente Northern California (KPNC), Oakland, CA, 94612, USA.
| | - Jie Yin
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California (KPNC), Oakland, CA 94612 USA
| | - Eric Jorgenson
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California (KPNC), Oakland, CA 94612 USA
| |
Collapse
|
18
|
Drake J, McMichael GO, Vornholt ES, Cresswell K, Williamson V, Chatzinakos C, Mamdani M, Hariharan S, Kendler KS, Kalsi G, Riley BP, Dozmorov M, Miles MF, Bacanu S, Vladimirov VI. Assessing the Role of Long Noncoding RNA in Nucleus Accumbens in Subjects With Alcohol Dependence. Alcohol Clin Exp Res 2020; 44:2468-2480. [PMID: 33067813 PMCID: PMC7756309 DOI: 10.1111/acer.14479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Long noncoding RNA (lncRNA) have been implicated in the etiology of alcohol use. Since lncRNA provide another layer of complexity to the transcriptome, assessing their expression in the brain is the first critical step toward understanding lncRNA functions in alcohol use and addiction. Thus, we sought to profile lncRNA expression in the nucleus accumbens (NAc) in a large postmortem alcohol brain sample. METHODS LncRNA and protein-coding gene (PCG) expressions in the NAc from 41 subjects with alcohol dependence (AD) and 41 controls were assessed via a regression model. Weighted gene coexpression network analysis was used to identify lncRNA and PCG networks (i.e., modules) significantly correlated with AD. Within the significant modules, key network genes (i.e., hubs) were also identified. The lncRNA and PCG hubs were correlated via Pearson correlations to elucidate the potential biological functions of lncRNA. The lncRNA and PCG hubs were further integrated with GWAS data to identify expression quantitative trait loci (eQTL). RESULTS At Bonferroni adj. p-value ≤ 0.05, we identified 19 lncRNA and 5 PCG significant modules, which were enriched for neuronal and immune-related processes. In these modules, we further identified 86 and 315 PCG and lncRNA hubs, respectively. At false discovery rate (FDR) of 10%, the correlation analyses between the lncRNA and PCG hubs revealed 3,125 positive and 1,860 negative correlations. Integration of hubs with genotype data identified 243 eQTLs affecting the expression of 39 and 204 PCG and lncRNA hubs, respectively. CONCLUSIONS Our study identified lncRNA and gene networks significantly associated with AD in the NAc, coordinated lncRNA and mRNA coexpression changes, highlighting potentially regulatory functions for the lncRNA, and our genetic (cis-eQTL) analysis provides novel insights into the etiological mechanisms of AD.
Collapse
Affiliation(s)
- John Drake
- From the Center for Integrative Life Sciences Education (JD)Virginia Commonwealth UniversityRichmondVirginia
| | - Gowon O. McMichael
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
| | - Eric Sean Vornholt
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
| | - Kellen Cresswell
- Department of Biostatistics(KC, MD)Virginia Commonwealth UniversityRichmondVirginia
| | - Vernell Williamson
- Department of Pathology(VW)Virginia Commonwealth UniversityRichmondVirginia
| | - Chris Chatzinakos
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
| | - Mohammed Mamdani
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
| | - Siddharth Hariharan
- Summer Research Fellowship(SH)School of MedicineVirginia Commonwealth UniversityRichmondVirginia
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Psychiatry(KSK, BPR, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Human and Molecular Genetics(KSK, BPR)Virginia Commonwealth UniversityRichmondVirginia
| | - Gursharan Kalsi
- Department of Social, Genetic and Developmental Psychiatry(GK)Institute of PsychiatryLondonUK
| | - Brien P. Riley
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Psychiatry(KSK, BPR, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Human and Molecular Genetics(KSK, BPR)Virginia Commonwealth UniversityRichmondVirginia
| | - Mikhail Dozmorov
- Department of Biostatistics(KC, MD)Virginia Commonwealth UniversityRichmondVirginia
| | - Michael F. Miles
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Pharmacology and Toxicology(MFM)Virginia Commonwealth UniversityRichmondVirginia
| | - Silviu‐Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Psychiatry(KSK, BPR, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
| | - Vladimir I. Vladimirov
- Virginia Institute for Psychiatric and Behavioral Genetics(GOM, ESV, CC, MM, KSK, BPR, MFM, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Department of Psychiatry(KSK, BPR, S‐AB, VIV)Virginia Commonwealth UniversityRichmondVirginia
- Center for Biomarker Research and Personalized Medicine(VIV)Virginia Commonwealth UniversityRichmondVirginia
- Lieber Institute for Brain Development(VIV)Johns Hopkins UniversityBaltimoreMaryland
| |
Collapse
|
19
|
Qu L, Wang L, He F, Han Y, Yang L, Wang MD, Zhu H. The Landscape of Micro-Inversions Provide Clues for Population Genetic Analysis of Humans. Interdiscip Sci 2020; 12:499-514. [PMID: 32929667 PMCID: PMC7658078 DOI: 10.1007/s12539-020-00392-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 11/04/2022]
Abstract
Background Variations in the human genome have been studied extensively. However, little is known about the role of micro-inversions (MIs), generally defined as small (< 100 bp) inversions, in human evolution, diversity, and health. Depicting the pattern of MIs among diverse populations is critical for interpreting human evolutionary history and obtaining insight into genetic diseases. Results In this paper, we explored the distribution of MIs in genomes from 26 human populations and 7 nonhuman primate genomes and analyzed the phylogenetic structure of the 26 human populations based on the MIs. We further investigated the functions of the MIs located within genes associated with human health. With hg19 as the reference genome, we detected 6968 MIs among the 1937 human samples and 24,476 MIs among the 7 nonhuman primate genomes. The analyses of MIs in human genomes showed that the MIs were rarely located in exonic regions. Nonhuman primates and human populations shared only 82 inverted alleles, and Africans had the most inverted alleles in common with nonhuman primates, which was consistent with the “Out of Africa” hypothesis. The clustering of MIs among the human populations also coincided with human migration history and ancestral lineages. Conclusions We propose that MIs are potential evolutionary markers for investigating population dynamics. Our results revealed the diversity of MIs in human populations and showed that they are essential to construct human population relationships and have a potential effect on human health. Electronic supplementary material The online version of this article (10.1007/s12539-020-00392-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li Qu
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, 30332, USA
| | - Luotong Wang
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - Feifei He
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - Yilun Han
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - Longshu Yang
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - May D Wang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, 30332, USA
| | - Huaiqiu Zhu
- State Key Laboratory for Turbulence and Complex Systems and Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China. .,Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, 30332, USA. .,Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| |
Collapse
|
20
|
Lyons K, Le LC, Pham YTH, Borron C, Park JY, Tran CTD, Tran TV, Tran HTT, Vu KT, Do CD, Pelucchi C, La Vecchia C, Zgibor J, Boffetta P, Luu HN. Gastric cancer: epidemiology, biology, and prevention: a mini review. Eur J Cancer Prev 2020; 28:397-412. [PMID: 31386635 DOI: 10.1097/cej.0000000000000480] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most common causes of cancer-related mortality worldwide. The objective of this article is to review the epidemiology and biology of gastric cancer risk. This literature review explores the biological, clinical, and environmental factors that influence the rates of this disease and discuss the different intervention methods that may not only increase the awareness of gastric cancer but also increase screening in efforts to reduce the risk of gastric cancer. Helicobacter pylori infection is the primary risk factor for gastric cancer. Additional risk factors include geographical location, age, sex, smoking, socioeconomic status, dietary intake, and genetics. Primary and secondary prevention strategies such as dietary modifications and screenings are important measures for reducing the risk of gastric cancer. Interventions, such as H. pylori eradication through chemoprevention trials, have shown some potential as a preventative strategy. Although knowledge about gastric cancer risk has greatly increased, future research is warranted on the differentiation of gastric cancer epidemiology by subsite and exploring the interactions between H. pylori infection, genetics, and environmental factors. Better understanding of these relationships can help researchers determine the most effective intervention strategies for reducing the risk of this disease.
Collapse
Affiliation(s)
- Kiara Lyons
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida
| | - Linh C Le
- VinUniversity Project-Health Sciences.,Vinmec Healthcare System
| | | | - Claire Borron
- Icahn School of Medicine, Mount Sinai School of Medicine, Tisch Cancer Institute, New York City, New York
| | - Jong Y Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Chi T D Tran
- Vietnam Colorectal Cancer and Polyp Research Program, Vinmec Healthcare System
| | - Thuan V Tran
- Vietnam National Cancer Hospital.,Vietnam National Cancer Institute
| | - Huong T-T Tran
- Vietnam National Cancer Hospital.,Vietnam National Cancer Institute
| | - Khanh T Vu
- Department of Gastroenterology, Bach Mai Hospital
| | - Cuong D Do
- Department of Infectious Disease, Bach Mai Hospital, Hanoi, Vietnam
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Janice Zgibor
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida
| | - Paolo Boffetta
- Icahn School of Medicine, Mount Sinai School of Medicine, Tisch Cancer Institute, New York City, New York
| | - Hung N Luu
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health.,Division of Cancer Control and Population Sciences, Hillman Cancer Canter, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
21
|
Schmitt RE, Messick MR, Shell BC, Dunbar EK, Fang H, Shelton KL, Venton BJ, Pletcher SD, Grotewiel M. Dietary yeast influences ethanol sedation in Drosophila via serotonergic neuron function. Addict Biol 2020; 25:e12779. [PMID: 31169340 DOI: 10.1111/adb.12779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 03/23/2019] [Accepted: 05/02/2019] [Indexed: 01/10/2023]
Abstract
Abuse of alcohol is a major clinical problem with far-reaching health consequences. Understanding the environmental and genetic factors that contribute to alcohol-related behaviors is a potential gateway for developing novel therapeutic approaches for patients that abuse the drug. To this end, we have used Drosophila melanogaster as a model to investigate the effect of diet, an environmental factor, on ethanol sedation. Providing flies with diets high in yeast, a routinely used component of fly media, increased their resistance to ethanol sedation. The yeast-induced resistance to ethanol sedation occurred in several different genetic backgrounds, was observed in males and females, was elicited by yeast from different sources, was readily reversible, and was associated with increased nutrient intake as well as decreased internal ethanol levels. Inhibition of serotonergic neuron function using multiple independent genetic manipulations blocked the effect of yeast supplementation on ethanol sedation, nutrient intake, and internal ethanol levels. Our results demonstrate that yeast is a critical dietary component that influences ethanol sedation in flies and that serotonergic signaling is required for the effect of dietary yeast on nutrient intake, ethanol uptake/elimination, and ethanol sedation. Our studies establish the fly as a model for diet-induced changes in ethanol sedation and raise the possibility that serotonin might mediate the effect of diet on alcohol-related behavior in other species.
Collapse
Affiliation(s)
- Rebecca E. Schmitt
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Monica R. Messick
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Brandon C. Shell
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Ellyn K. Dunbar
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Huai‐Fang Fang
- Department of Chemistry and Neuroscience Graduate Program University of Virginia Charlottesville VA USA
| | - Keith L. Shelton
- Department of Pharmacology and Toxicology Virginia Commonwealth University Richmond VA USA
| | - B. Jill Venton
- Department of Chemistry and Neuroscience Graduate Program University of Virginia Charlottesville VA USA
| | - Scott D. Pletcher
- Department of Molecular and Integrative Physiology and Geriatrics Center University of Michigan Ann Arbor MI USA
| | - Mike Grotewiel
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
- Virginia Commonwealth University Alcohol Research Center Richmond VA USA
| |
Collapse
|
22
|
Dash GF, Davis CN, Martin NG, Statham DJ, Lynskey MT, Slutske WS. High-Intensity Drinking in Adult Australian Twins. Alcohol Clin Exp Res 2020; 44:522-531. [PMID: 31943258 DOI: 10.1111/acer.14262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Many adult drinkers consume far beyond the binge threshold. This "high-intensity drinking" (HID), defined as 2 (HID-2) and 3 (HID-3) times the binge threshold, is of public health interest due to its role in acute alcohol-related harms. Research on HID has mostly been limited to college-aged young adults, focused on contextual factors, and neglected the potential role of genetic influences on the propensity to engage in HID. METHODS Structured diagnostic interviews assessing past-year alcohol involvement were conducted with 3,785 individuals (1,365 men, 2,420 women; Mage = 32, range = 21 to 46), including 3,314 twins and 471 nontwin siblings from the Australian Twin Registry. Multinomial logistic regression analyses were conducted to compare HID-2 and HID-3 to binge drinking on demographic correlates, drinking characteristics, and drinking-related consequences. Biometric modeling was conducted to estimate the role of genetic, common, and individual-specific environmental factors in HID propensity. RESULTS Among past-year drinkers, the prevalence of HID-2 and HID-3 was both 22%, with men disproportionally represented. The frequencies of drinking, intoxication, and binge drinking significantly increased across the heavier drinking categories, which also evidenced higher average consumption quantities and higher rates of alcohol-related consequences. The propensity to engage in HID was significantly heritable (A = 37% [95% CI: 28 to 46%]), with individual-specific environmental influences accounting for the remainder of the variance. CONCLUSIONS This study convincingly demonstrates that HID is not restricted to college-aged young adults, but also can be highly prevalent among those of working age, and that the propensity to engage in HID is partially explained by genetic influences.
Collapse
Affiliation(s)
- Genevieve F Dash
- From the, Department of Psychological Sciences, (GFD, CND, WSS), University of Missouri- Columbia, Columbia, Missouri
| | - Christal N Davis
- From the, Department of Psychological Sciences, (GFD, CND, WSS), University of Missouri- Columbia, Columbia, Missouri
| | - Nicholas G Martin
- Queensland Institute of Medical Research (QIMR) Berghofer, (NGM), Brisbane, QLD, Australia
| | - Dixie J Statham
- Department of Psychology, (DJS), Federation University, Ballarat, Vic., Australia
| | - Michael T Lynskey
- Department of Addictions, (MTL), King's College London Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Wendy S Slutske
- From the, Department of Psychological Sciences, (GFD, CND, WSS), University of Missouri- Columbia, Columbia, Missouri
| |
Collapse
|
23
|
McQuire C, Daniel R, Hurt L, Kemp A, Paranjothy S. The causal web of foetal alcohol spectrum disorders: a review and causal diagram. Eur Child Adolesc Psychiatry 2020; 29:575-594. [PMID: 30648224 PMCID: PMC7250957 DOI: 10.1007/s00787-018-1264-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022]
Abstract
Foetal alcohol spectrum disorders (FASDs) are a leading cause of developmental disability. Prenatal alcohol use is the sole necessary cause of FASD, but it is not always sufficient. Multiple factors influence a child's susceptibility to FASD following prenatal alcohol exposure. Much of the FASD risk factor literature has been limited to discussions of association, rather than causation. While knowledge of predictor variables is important for identifying who is most at risk of FASD and for targeting interventions, causal knowledge is important for identifying effective mechanisms for prevention and intervention programmes. We conducted a systematic search and narrative synthesis of the evidence and used this to create a causal diagram (directed acyclic graph; DAG) to describe the causal pathways to FASD. Our results show that the aetiology of FASD is multifaceted and complex. FASD risk is determined by a range of lifestyle, sociodemographic, maternal, social, gestational, and genetic factors. The causal diagram that we present in this review provides a comprehensive summary of causal risk factors for FASD and can be used as a tool to inform data collection and statistical modelling strategies to minimise bias in future studies of FASD.
Collapse
Affiliation(s)
- Cheryl McQuire
- Population Health Sciences, Bristol Medical School, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK.
| | - R. Daniel
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - L. Hurt
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - A. Kemp
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - S. Paranjothy
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| |
Collapse
|
24
|
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
|
25
|
Gelernter J, Sun N, Polimanti R, Pietrzak RH, Levey DF, Lu Q, Hu Y, Li B, Radhakrishnan K, Aslan M, Cheung KH, Li Y, Rajeevan N, Sayward F, Harrington K, Chen Q, Cho K, Honerlaw J, Pyarajan S, Lencz T, Quaden R, Shi Y, Hunter-Zinck H, Gaziano JM, Kranzler HR, Concato J, Zhao H, Stein MB. Genome-wide Association Study of Maximum Habitual Alcohol Intake in >140,000 U.S. European and African American Veterans Yields Novel Risk Loci. Biol Psychiatry 2019; 86:365-376. [PMID: 31151762 PMCID: PMC6919570 DOI: 10.1016/j.biopsych.2019.03.984] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Habitual alcohol use can be an indicator of alcohol dependence, which is associated with a wide range of serious health problems. METHODS We completed a genome-wide association study in 126,936 European American and 17,029 African American subjects in the Veterans Affairs Million Veteran Program for a quantitative phenotype based on maximum habitual alcohol consumption. RESULTS ADH1B, on chromosome 4, was the lead locus for both populations: for the European American sample, rs1229984 (p = 4.9 × 10-47); for African American, rs2066702 (p = 2.3 × 10-12). In the European American sample, we identified three additional genome-wide-significant maximum habitual alcohol consumption loci: on chromosome 17, rs77804065 (p = 1.5 × 10-12), at CRHR1 (corticotropin-releasing hormone receptor 1); the protein product of this gene is involved in stress and immune responses; and on chromosomes 8 and 10. European American and African American samples were then meta-analyzed; the associated region at CRHR1 increased in significance to 1.02 × 10-13, and we identified two additional genome-wide significant loci, FGF14 (p = 9.86 × 10-9) (chromosome 13) and a locus on chromosome 11. Besides ADH1B, none of the five loci have prior genome-wide significant support. Post-genome-wide association study analysis identified genetic correlation to other alcohol-related traits, smoking-related traits, and many others. Replications were observed in UK Biobank data. Genetic correlation between maximum habitual alcohol consumption and alcohol dependence was 0.87 (p = 4.78 × 10-9). Enrichment for cell types included dopaminergic and gamma-aminobutyric acidergic neurons in midbrain, and pancreatic delta cells. CONCLUSIONS The present study supports five novel alcohol-use risk loci, with particularly strong statistical support for CRHR1. Additionally, we provide novel insight regarding the biology of harmful alcohol use.
Collapse
Affiliation(s)
- Joel Gelernter
- Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| | - Ning Sun
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Renato Polimanti
- Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Robert H Pietrzak
- Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Daniel F Levey
- Psychiatry Service, VA Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Qiongshi Lu
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Yiming Hu
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Boyang Li
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Krishnan Radhakrishnan
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut
| | - Mihaela Aslan
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Kei-Hoi Cheung
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Emergency Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Yuli Li
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Yale Center for Medical Informatics, Yale University School of Medicine, New Haven, Connecticut
| | - Nallakkandi Rajeevan
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Yale Center for Medical Informatics, Yale University School of Medicine, New Haven, Connecticut
| | - Fred Sayward
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Yale Center for Medical Informatics, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly Harrington
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts; Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts
| | - Quan Chen
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jacqueline Honerlaw
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts
| | - Saiju Pyarajan
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Todd Lencz
- Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, New York; Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, New York; Department of Psychiatry, Division of Research, The Zucker Hillside Hospital Division of Northwell Health, Glen Oaks, New York; Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Rachel Quaden
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts
| | - Yunling Shi
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts
| | - Haley Hunter-Zinck
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Henry R Kranzler
- Veterans Integrated Services Networks (VISN) 4 Mental Illness Research, Education and Clinical Center, Crescenz VA Medical Center, Philadelphia, Pennsylvania; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - John Concato
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Hongyu Zhao
- Veterans Affairs (VA) Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut; Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut
| | - Murray B Stein
- Psychiatry Service, VA San Diego Healthcare System, San Diego, California; Department of Psychiatry, University of California San Diego, La Jolla, California.
| |
Collapse
|
26
|
Affiliation(s)
- Howard J Edenberg
- Departments of Biochemistry and Molecular Biology and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
27
|
Cornelis MC. Genetic determinants of beverage consumption: Implications for nutrition and health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 89:1-52. [PMID: 31351524 PMCID: PMC7047661 DOI: 10.1016/bs.afnr.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Beverages make important contributions to nutritional intake and their role in health has received much attention. This review focuses on the genetic determinants of common beverage consumption and how research in this field is contributing insight to what and how much we consume and why this genetic knowledge matters from a research and public health perspective. The earliest efforts in gene-beverage behavior mapping involved genetic linkage and candidate gene analysis but these approaches have been largely replaced by genome-wide association studies (GWAS). GWAS have identified biologically plausible loci underlying alcohol and coffee drinking behavior. No GWAS has identified variants specifically associated with consumption of tea, juice, soda, wine, beer, milk or any other common beverage. Thus far, GWAS highlight an important behavior-reward component (as opposed to taste) to beverage consumption which may serve as a potential barrier to dietary interventions. Loci identified have been used in Mendelian randomization and gene×beverage interaction analysis of disease but results have been mixed. This research is necessary as it informs the clinical relevance of SNP-beverage associations and thus genotype-based personalized nutrition, which is gaining interest in the commercial and public health sectors.
Collapse
Affiliation(s)
- Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
28
|
Kranzler HR, Zhou H, Kember RL, Vickers Smith R, Justice AC, Damrauer S, Tsao PS, Klarin D, Baras A, Reid J, Overton J, Rader DJ, Cheng Z, Tate JP, Becker WC, Concato J, Xu K, Polimanti R, Zhao H, Gelernter J. Genome-wide association study of alcohol consumption and use disorder in 274,424 individuals from multiple populations. Nat Commun 2019; 10:1499. [PMID: 30940813 PMCID: PMC6445072 DOI: 10.1038/s41467-019-09480-8] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/06/2019] [Indexed: 12/21/2022] Open
Abstract
Alcohol consumption level and alcohol use disorder (AUD) diagnosis are moderately heritable traits. We conduct genome-wide association studies of these traits using longitudinal Alcohol Use Disorder Identification Test-Consumption (AUDIT-C) scores and AUD diagnoses in a multi-ancestry Million Veteran Program sample (N = 274,424). We identify 18 genome-wide significant loci: 5 associated with both traits, 8 associated with AUDIT-C only, and 5 associated with AUD diagnosis only. Polygenic Risk Scores (PRS) for both traits are associated with alcohol-related disorders in two independent samples. Although a significant genetic correlation reflects the overlap between the traits, genetic correlations for 188 non-alcohol-related traits differ significantly for the two traits, as do the phenotypes associated with the traits' PRS. Cell type group partitioning heritability enrichment analyses also differentiate the two traits. We conclude that, although heavy drinking is a key risk factor for AUD, it is not a sufficient cause of the disorder.
Collapse
Affiliation(s)
- Henry R Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA.
| | - Hang Zhou
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Rachel L Kember
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Rachel Vickers Smith
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
- University of Louisville School of Nursing, Louisville, KY, 40202, USA
| | - Amy C Justice
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - Scott Damrauer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Derek Klarin
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - Jeffrey Reid
- Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - John Overton
- Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - Daniel J Rader
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Zhongshan Cheng
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Janet P Tate
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - William C Becker
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - John Concato
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Ke Xu
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Renato Polimanti
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Hongyu Zhao
- Yale School of Medicine, New Haven, CT, 06511, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - Joel Gelernter
- Yale School of Medicine, New Haven, CT, 06511, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, 06516, USA
| |
Collapse
|
29
|
Sanchez-Roige S, Fontanillas P, Elson SL, Gray JC, de Wit H, Davis LK, MacKillop J, Palmer AA. Genome-wide association study of alcohol use disorder identification test (AUDIT) scores in 20 328 research participants of European ancestry. Addict Biol 2019; 24:121-131. [PMID: 29058377 PMCID: PMC6988186 DOI: 10.1111/adb.12574] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022]
Abstract
Genetic factors contribute to the risk for developing alcohol use disorder (AUD). In collaboration with the genetics company 23andMe, Inc., we performed a genome-wide association study of the alcohol use disorder identification test (AUDIT), an instrument designed to screen for alcohol misuse over the past year. Our final sample consisted of 20 328 research participants of European ancestry (55.3% females; mean age = 53.8, SD = 16.1) who reported ever using alcohol. Our results showed that the 'chip-heritability' of AUDIT score, when treated as a continuous phenotype, was 12%. No loci reached genome-wide significance. The gene ADH1C, which has been previously implicated in AUD, was among our most significant associations (4.4 × 10-7 ; rs141973904). We also detected a suggestive association on chromosome 1 (2.1 × 10-7 ; rs182344113) near the gene KCNJ9, which has been implicated in mouse models of high ethanol drinking. Using linkage disequilibrium score regression, we identified positive genetic correlations between AUDIT score, high alcohol consumption and cigarette smoking. We also observed an unexpected positive genetic correlation between AUDIT and educational attainment and additional unexpected negative correlations with body mass index/obesity and attention-deficit/hyperactivity disorder. We conclude that conducting a genetic study using responses to an online questionnaire in a population not ascertained for AUD may represent a cost-effective strategy for elucidating aspects of the etiology of AUD.
Collapse
Affiliation(s)
- Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | | | | | | | - Joshua C. Gray
- Center for Deployment Psychology, Uniformed Services University, Bethesda, MD, 20814
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Lea K. Davis
- Vanderbilt Genetics Institute; Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University/St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada; Homewood Research Institute, Guelph, ON N1E 6K9, Canada
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
30
|
Brick LA, Keller MC, Knopik VS, McGeary JE, Palmer RHC. Shared additive genetic variation for alcohol dependence among subjects of African and European ancestry. Addict Biol 2019; 24:132-144. [PMID: 29178570 PMCID: PMC6312725 DOI: 10.1111/adb.12578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 09/05/2017] [Accepted: 10/15/2017] [Indexed: 02/01/2023]
Abstract
Alcohol dependence (AD) affects individuals from all racial/ethnic groups, and previous research suggests that there is considerable variation in AD risk between and among various ancestrally defined groups in the United States. Although the reasons for these differences are likely due in part to contributions of complex sociocultural factors, limited research has attempted to examine whether similar genetic variation plays a role across ancestral groups. Using a pooled sample of individuals of African and European ancestry (AA/EA) obtained through data shared within the Database for Genotypes and Phenotypes, we estimated the extent to which additive genetic similarity for AD between AA and EAs using common single nucleotide polymorphisms overlapped across the two populations. AD was represented as a factor score by using Diagnostic and Statistical Manual dependence criteria, and genetic data were imputed by using the 1000 Genomes Reference Panel. Analyses revealed a significant single nucleotide polymorphism-based heritability of 17 percent (SE = 5) in EAs and 24 percent (SE = 15) in AAs. Further, a significant genetic correlation of 0.77 (SE = 0.46) suggests that the allelic architecture influencing the AD factor for EAs and AAs is largely similar across the two populations. Analyses indicated that investigating the genetic underpinnings of alcohol dependence in different ethnic groups may serve to highlight core etiological factors common to both groups and unique etiological factors specific to each ethnic group.
Collapse
Affiliation(s)
- Leslie A. Brick
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Matthew C. Keller
- Institute for Behavior Genetics, department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, Colorado
| | - Valerie S. Knopik
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, Rhode Island
| | - John E. McGeary
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, Rhode Island
- Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Rohan H. C. Palmer
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, Rhode Island
- Behavior Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, Georgia
| |
Collapse
|
31
|
Yang M, Zhang Y, Ren J. ALDH2 Polymorphism and Ethanol Consumption: A Genetic-Environmental Interaction in Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:229-236. [DOI: 10.1007/978-981-13-6260-6_14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
32
|
Jiménez-Jiménez FJ, Gómez-Tabales J, Alonso-Navarro H, Zurdo M, Turpín-Fenoll L, Millán-Pascual J, Adeva-Bartolomé T, Cubo E, Navacerrada F, Rojo-Sebastián A, Rubio L, Díez-Fairén M, Pastor P, Calleja M, Plaza-Nieto JF, Pilo-de-la-Fuente B, Arroyo-Solera M, García-Albea E, Agúndez JAG, García-Martín E. Association Between the rs1229984 Polymorphism in the Alcohol Dehydrogenase 1B Gene and Risk for Restless Legs Syndrome. Sleep 2018; 40:4554483. [PMID: 29045753 DOI: 10.1093/sleep/zsx174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background/Objectives Several studies have raised the possibility of an association between alcohol consumption and the risk of developing restless legs syndrome (RLS). Moreover, an important percentage of patients under alcohol detoxification therapy develop RLS symptoms that fulfil the criteria for idiopathic RLS during alcohol withdrawal. We have aimed to establish the possible association between two common single nucleotide polymorphisms (SNPs) in the alcohol-dehydrogenase 1B (ADH1B) gene and the risk for RLS. Methods We studied, using specific TaqMan assays, the genotype and allelic variant frequencies of ADH1B rs1229984 and ADH1B rs6413413 SNPs in 205 RLS patients and 505 gender-matched healthy controls. Results The sum of the frequencies of rs1229984CT and rs1229984TT genotypes, as well as the frequency of the rs1229984T allelic variant, was significantly higher in RLS patients than in controls, both in the whole group and in females. The frequencies of genotypes and allelic variants of the rs6413413 SNP were similar between the two groups. RLS patients with the rs1229984CT genotype were younger, and those with the rs122984TT genotype older, at onset of RLS symptoms than those with the rs1229984CC genotype. None of the studied SNPs were related either with positivity of family history for RLS or with RLS severity. Conclusions These results suggest an association between rs1229984 SNP and the risk for RLS.
Collapse
Affiliation(s)
- Félix Javier Jiménez-Jiménez
- Section of Neurology, Hospital Universitario del Sureste, Spain.,Department of Medicine-Neurology, Hospital "Príncipe de Asturias," Universidad de Alcalá, Spain
| | | | - Hortensia Alonso-Navarro
- Section of Neurology, Hospital Universitario del Sureste, Spain.,Department of Medicine-Neurology, Hospital "Príncipe de Asturias," Universidad de Alcalá, Spain
| | - Martín Zurdo
- Section of Neurology, Hospital Virgen del Puerto, Spain
| | | | | | | | - Esther Cubo
- Section of Neurology, Hospital Universitario de Burgos, Spain
| | | | - Ana Rojo-Sebastián
- Department of Medicine-Neurology, Hospital "Príncipe de Asturias," Universidad de Alcalá, Spain
| | - Lluisa Rubio
- Department of Medicine-Neurology, Hospital "Príncipe de Asturias," Universidad de Alcalá, Spain
| | - Mónica Díez-Fairén
- Fundació per la Recerca Biomèdica i Social Mútua de Terrassa, Spain.,Department of Neurology, Memory and Movement Disorders Units, Hospital Universitari Mutua de Terrassa, Spain
| | - Pau Pastor
- Fundació per la Recerca Biomèdica i Social Mútua de Terrassa, Spain.,Department of Neurology, Memory and Movement Disorders Units, Hospital Universitari Mutua de Terrassa, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain.,Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, Universidad de Navarra, Spain
| | - Marisol Calleja
- Section of Neurology, Hospital Universitario del Sureste, Spain
| | | | | | | | - Esteban García-Albea
- Department of Medicine-Neurology, Hospital "Príncipe de Asturias," Universidad de Alcalá, Spain
| | | | | |
Collapse
|
33
|
Edenberg HJ, McClintick JN. Alcohol Dehydrogenases, Aldehyde Dehydrogenases, and Alcohol Use Disorders: A Critical Review. Alcohol Clin Exp Res 2018; 42:2281-2297. [PMID: 30320893 PMCID: PMC6286250 DOI: 10.1111/acer.13904] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/07/2018] [Indexed: 12/20/2022]
Abstract
Alcohol use disorders (AUDs) are complex traits, meaning that variations in many genes contribute to the risk, as does the environment. Although the total genetic contribution to risk is substantial, most individual variations make only very small contributions. By far the strongest contributors are functional variations in 2 genes involved in alcohol (ethanol [EtOH]) metabolism. A functional variant in alcohol dehydrogenase 1B (ADH1B) is protective in people of European and Asian descent, and a different functional variant in the same gene is protective in those of African descent. A strongly protective variant in aldehyde dehydrogenase 2 (ALDH2) is essentially only found in Asians. This highlights the need to study a wide range of populations. The likely mechanism of protection against heavy drinking and AUDs in both cases is alteration in the rate of metabolism of EtOH that at least transiently elevates acetaldehyde. Other ADH and ALDH variants, including functional variations in ADH1C, have also been implicated in affecting drinking behavior and risk for alcoholism. The pattern of linkage disequilibrium in the ADH region and the differences among populations complicate analyses, particularly of regulatory variants. This critical review focuses upon the ADH and ALDH genes as they affect AUDs.
Collapse
Affiliation(s)
- Howard J. Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Jeanette N. McClintick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
34
|
Justice AC, Smith RV, Tate JP, McGinnis K, Xu K, Becker WC, Lee KY, Lynch K, Sun N, Concato J, Fiellin DA, Zhao H, Gelernter J, Kranzler HR. AUDIT-C and ICD codes as phenotypes for harmful alcohol use: association with ADH1B polymorphisms in two US populations. Addiction 2018; 113:2214-2224. [PMID: 29972609 PMCID: PMC6226338 DOI: 10.1111/add.14374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/31/2018] [Accepted: 06/28/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS Longitudinal electronic health record (EHR) data offer a large-scale, untapped source of phenotypical information on harmful alcohol use. Using established, alcohol-associated variants in the gene that encodes the enzyme alcohol dehydrogenase 1B (ADH1B) as criterion standards, we compared the individual and combined validity of three longitudinal EHR-based phenotypes of harmful alcohol use: Alcohol Use Disorders Identification Test-Consumption (AUDIT-C) trajectories; mean age-adjusted AUDIT-C; and diagnoses of alcohol use disorder (AUD). DESIGN With longitudinal EHR data from the Million Veteran Program (MVP) linked to genetic data, we used two population-specific polymorphisms in ADH1B that are associated strongly with AUD in African Americans (AAs) and European Americans (EAs): rs2066702 (Arg369Cys, AAs) and rs1229984 (Arg48His, EAs) as criterion measures. SETTING United States Department of Veterans Affairs Healthcare System. PARTICIPANTS A total of 167 721 veterans (57 677 AAs and 110 044 EAs; 92% male, mean age = 63 years) took part in this study. Data were collected from 1 October 2007 to 1 May 2017. MEASUREMENTS Using all AUDIT-C scores and AUD diagnostic codes recorded in the EHR, we calculated age-adjusted mean AUDIT-C values, longitudinal statistical trajectories of AUDIT-C scores and ICD-9/10 diagnostic groupings for AUD. FINDINGS A total of 19 793 AAs (34.3%) had one or two minor alleles at rs2066702 [minor allele frequency (MAF) = 0.190] and 6933 EAs (6.3%) had one or two minor alleles at rs1229984 (MAF = 0.032). In both populations, trajectories and age-adjusted mean AUDIT-C were correlated (r = 0.90) but, when considered separately, highest score (8+ versus 0) of age-adjusted mean AUDIT-C demonstrated a stronger association with the ADH1B variants [adjusted odds ratio (aOR) 0.54 in AAs and 0.37 in AAs] than did the highest trajectory (aOR 0.71 in AAs and 0.53 in EAs); combining AUDIT-C metrics did not improve discrimination. When age-adjusted mean AUDIT-C score and AUD diagnoses were considered together, age-adjusted mean AUDIT-C (8+ versus 0) was associated with lower odds of having the ADH1B minor allele than were AUD diagnostic codes: aOR = 0.59 versus 0.86 in AAs and 0.48 versus 0.68 in EAs. These independent associations combine to yield an even lower aOR of 0.51 for AAs and 0.33 for EAs. CONCLUSIONS The age-adjusted mean AUDIT-C score is associated more strongly with genetic polymorphisms of known risk for alcohol use disorder than are longitudinal trajectories of AUDIT-C or AUD diagnostic codes. AUD diagnostic codes modestly enhance this association.
Collapse
Affiliation(s)
- Amy C. Justice
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516,Yale School of Public Health, New Haven CT 06515
| | - Rachel V. Smith
- University of Louisville School of Nursing, Louisville, KY 40202
| | | | - Kathleen McGinnis
- Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Ke Xu
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - William C. Becker
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Kuang-Yao Lee
- Department of Statistical Science, Temple University, Philadelphia, PA 19122
| | - Kevin Lynch
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA 19104,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Ning Sun
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - John Concato
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - David A. Fiellin
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - Hongyu Zhao
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - Joel Gelernter
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Henry R. Kranzler
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA 19104,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | | |
Collapse
|
35
|
Association between the missense alcohol dehydrogenase rs1229984T variant with the risk for Parkinson's disease in women. J Neurol 2018; 266:346-352. [PMID: 30483881 DOI: 10.1007/s00415-018-9136-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND/OBJECTIVE Several meta-analyses including retrospective case-control studies have shown that the risk of developing Parkinson's disease (PD) correlates inversely with alcohol consumption and (PD), although the results of prospective longitudinal studies are far from being conclusive. The reasons for this inverse association are not well-known. Because alcohol dehydrogenase is one of the most important alcohol-detoxification enzymes, we tried to replicate a putative association of the risk of developing PD with two missense gene variations affecting the alcohol dehydrogenase 1B (ADH1B) gene (one of them related with aversive effects to alcohol). METHODS In a cohort composed of 629 PD patients and 865 age- and gender-matched healthy individuals, we analyzed genotypes and allele frequencies for two common missense ADH1B single nucleotide polymorphisms (SNPs), namely rs1229984 (His48Arg) and rs6413413 (Thr60Ser) using specifically designed TaqMan assays. RESULTS The frequency of individuals carrying rs1229984T alleles in homozygosity or in heterozygosity was higher in PD than in controls in the whole study cohort (P < 0.001 and P = 0.005, respectively), and in women (P < 0.001 and P < 0.001, respectively). The genotypes for rs6413413 were similar in PD patients and control subjects. Age at onset of PD patients was not statistically related to rs1229984 or rs6413413 genotypes. CONCLUSIONS The missense variant rs1229984T is statistically associated with the risk of developing PD mainly in women, which could explain differences in alcohol consumption in this gender.
Collapse
|
36
|
Almli LM, Lori A, Meyers JL, Shin J, Fani N, Maihofer AX, Nievergelt CM, Smith AK, Mercer KB, Kerley K, Leveille JM, Feng H, Abu‐Amara D, Flory JD, Yehuda R, Marmar CR, Baker DG, Bradley B, Koenen KC, Conneely KN, Ressler KJ. Problematic alcohol use associates with sodium channel and clathrin linker 1 (SCLT1) in trauma-exposed populations. Addict Biol 2018; 23:1145-1159. [PMID: 29082582 DOI: 10.1111/adb.12569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 08/05/2017] [Accepted: 08/29/2017] [Indexed: 12/15/2022]
Abstract
Excessive alcohol use is extremely prevalent in the United States, particularly among trauma-exposed individuals. While several studies have examined genetic influences on alcohol use and related problems, this has not been studied in the context of trauma-exposed populations. We report results from a genome-wide association study of alcohol consumption and associated problems as measured by the alcohol use disorders identification test (AUDIT) in a trauma-exposed cohort. Results indicate a genome-wide significant association between total AUDIT score and rs1433375 [N = 1036, P = 2.61 × 10-8 (dominant model), P = 7.76 × 10-8 (additive model)], an intergenic single-nucleotide polymorphism located 323 kb upstream of the sodium channel and clathrin linker 1 (SCLT1) at 4q28. rs1433375 was also significant in a meta-analysis of two similar, but independent, cohorts (N = 1394, P = 0.0004), the Marine Resiliency Study and Systems Biology PTSD Biomarkers Consortium. Functional analysis indicated that rs1433375 was associated with SCLT1 gene expression and cortical-cerebellar functional connectivity measured via resting state functional magnetic resonance imaging. Together, findings suggest a role for sodium channel regulation and cerebellar functioning in alcohol use behavior. Identifying mechanisms underlying risk for problematic alcohol use in trauma-exposed populations is critical for future treatment and prevention efforts.
Collapse
Affiliation(s)
- Lynn M. Almli
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry State University of New York Downstate Medical Center Brooklyn NY USA
| | - Jaemin Shin
- Center for Advanced Brain Imaging Georgia State University/Georgia Institute of Technology Atlanta GA USA
| | - Negar Fani
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
| | - Adam X. Maihofer
- Department of Psychiatry University of California San Diego San Diego CA USA
- Veterans Affairs Center of Excellence for Stress and Mental Health San Diego USA
| | - Caroline M. Nievergelt
- Department of Psychiatry University of California San Diego San Diego CA USA
- Veterans Affairs Center of Excellence for Stress and Mental Health San Diego USA
| | - Alicia K. Smith
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
- Department of Gynecology and Obstetrics Emory University Atlanta GA USA
| | | | - Kimberly Kerley
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
| | - Jennifer M. Leveille
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
| | - Hao Feng
- Department of Human Genetics Emory University Atlanta GA USA
| | - Duna Abu‐Amara
- Steven and Alexandra Cohen Veterans Center for Posttraumatic Stress and Traumatic Brain Injury Department of Psychiatry, New York University New York NY USA
| | - Janine D. Flory
- Steven and Alexandra Cohen Veterans Center for Posttraumatic Stress and Traumatic Brain Injury Department of Psychiatry, New York University New York NY USA
- Department of Psychiatry MSSM/James J. Peters Veterans Administration Medical Center New York NY USA
| | - Rachel Yehuda
- Steven and Alexandra Cohen Veterans Center for Posttraumatic Stress and Traumatic Brain Injury Department of Psychiatry, New York University New York NY USA
- Department of Psychiatry MSSM/James J. Peters Veterans Administration Medical Center New York NY USA
| | - Charles R. Marmar
- Steven and Alexandra Cohen Veterans Center for Posttraumatic Stress and Traumatic Brain Injury Department of Psychiatry, New York University New York NY USA
| | - Dewleen G. Baker
- Department of Psychiatry University of California San Diego San Diego CA USA
- Veterans Affairs Center of Excellence for Stress and Mental Health San Diego USA
- Psychiatry Services VA San Diego Healthcare System San Diego CA USA
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
- Mental Health Service Line Department of Veterans Affairs Medical Center Atlanta GA USA
| | - Karestan C. Koenen
- Department of Epidemiology Harvard TH Chan School of Public Health Boston MA USA
| | | | - Kerry J. Ressler
- Department of Psychiatry and Behavioral Sciences Emory University Atlanta GA USA
- McLean Hospital Harvard Medical School Belmont MA USA
| |
Collapse
|
37
|
Iancu OD, Colville AM, Wilmot B, Searles R, Darakjian P, Zheng C, McWeeney S, Kawane S, Crabbe JC, Metten P, Oberbeck D, Hitzemann R. Gender-Specific Effects of Selection for Drinking in the Dark on the Network Roles of Coding and Noncoding RNAs. Alcohol Clin Exp Res 2018; 42:1454-1465. [PMID: 29786871 DOI: 10.1111/acer.13777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Transcriptional differences between heterogeneous stock mice and high drinking-in-the-dark selected mouse lines have previously been described based on microarray technology coupled with network-based analysis. The network changes were reproducible in 2 independent selections and largely confined to 2 distinct network modules; in contrast, differential expression appeared more specific to each selected line. This study extends these results by utilizing RNA-Seq technology, allowing evaluation of the relationship between genetic risk and transcription of noncoding RNA (ncRNA); we additionally evaluate sex-specific transcriptional effects of selection. METHODS Naïve mice (N = 24/group and sex) were utilized for gene expression analysis in the ventral striatum; the transcriptome was sequenced with the Illumina HiSeq platform. Differential gene expression and the weighted gene co-expression network analysis were implemented largely as described elsewhere, resulting in the identification of genes that change expression level or (co)variance structure. RESULTS Across both sexes, we detect selection effects on the extracellular matrix and synaptic signaling, although the identity of individual genes varies. A majority of nc RNAs cluster in a single module of relatively low density in both the male and female network. The most strongly differentially expressed transcript in both sexes was Gm22513, a small nuclear RNA with unknown function. Associated with selection, we also found a number of network hubs that change edge strength and connectivity. At the individual gene level, there are many sex-specific effects; however, at the annotation level, results are more concordant. CONCLUSIONS In addition to demonstrating sex-specific effects of selection on the transcriptome, the data point to the involvement of extracellular matrix genes as being associated with the binge drinking phenotype.
Collapse
Affiliation(s)
- Ovidiu Dan Iancu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Alex M Colville
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Beth Wilmot
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Robert Searles
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, Oregon
| | - Priscila Darakjian
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Christina Zheng
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Shannon McWeeney
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon
| | - Sunita Kawane
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon
| | - John C Crabbe
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon.,VA Portland Health Care System , Portland, Oregon
| | - Pamela Metten
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon.,VA Portland Health Care System , Portland, Oregon
| | - Denesa Oberbeck
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
38
|
Koulentaki M, Kouroumalis E. GABA A receptor polymorphisms in alcohol use disorder in the GWAS era. Psychopharmacology (Berl) 2018; 235:1845-1865. [PMID: 29721579 DOI: 10.1007/s00213-018-4918-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/18/2018] [Indexed: 12/11/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing, neuro-psychiatric illness of high prevalence and with a serious public health impact worldwide. It is complex and polygenic, with a heritability of about 50%, and influenced by environmental causal heterogeneity. Risk factors associated with its etiology have a genetic component. GABA (γ-aminobutyric acid) is a major inhibitory neurotransmitter in mammalian brain. GABAA receptors are believed to mediate some of the physiological and behavioral actions of alcohol. In this critical review, relevant genetic terms and type and methodology of the genetic studies are briefly explained. Postulated candidate genes that encode subunits of GABAA receptors, with all the reported SNPs, are presented. Genetic studies and meta-analyses examining polymorphisms of the GABAA receptor and their association with AUD predisposition are presented. The data are critically examined with reference to recent GWAS studies that failed to show relations between GABAA receptors and AUD. Restrictions and perspectives of the different findings are discussed.
Collapse
Affiliation(s)
- Mairi Koulentaki
- Alcohology Research Laboratory, Medical School, University of Crete, 71500, Heraklion, Crete, Greece.,Department of Gastroenterology, University Hospital Heraklion, 71500, Heraklion, Crete, Greece
| | - Elias Kouroumalis
- Department of Gastroenterology, University Hospital Heraklion, 71500, Heraklion, Crete, Greece.
| |
Collapse
|
39
|
Bogdan R, Baranger DAA, Agrawal A. Polygenic Risk Scores in Clinical Psychology: Bridging Genomic Risk to Individual Differences. Annu Rev Clin Psychol 2018; 14:119-157. [PMID: 29579395 PMCID: PMC7772939 DOI: 10.1146/annurev-clinpsy-050817-084847] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomewide association studies (GWASs) across psychiatric phenotypes have shown that common genetic variants generally confer risk with small effect sizes (odds ratio < 1.1) that additively contribute to polygenic risk. Summary statistics derived from large discovery GWASs can be used to generate polygenic risk scores (PRS) in independent, target data sets to examine correlates of polygenic disorder liability (e.g., does genetic liability to schizophrenia predict cognition?). The intuitive appeal and generalizability of PRS have led to their widespread use and new insights into mechanisms of polygenic liability. However, when currently applied across traits they account for small amounts of variance (<3%), are relatively uninformative for clinical treatment, and, in isolation, provide no insight into molecular mechanisms. Larger GWASs are needed to increase the precision of PRS, and novel approaches integrating various data sources (e.g., multitrait analysis of GWASs) may improve the utility of current PRS.
Collapse
Affiliation(s)
- Ryan Bogdan
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - David A A Baranger
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
40
|
Gelernter J, Zhou H, Nuñez YZ, Mutirangura A, Malison RT, Kalayasiri R. Genomewide Association Study of Alcohol Dependence and Related Traits in a Thai Population. Alcohol Clin Exp Res 2018; 42:861-868. [PMID: 29460428 DOI: 10.1111/acer.13614] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/14/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alcohol use (both quantity and dependence) is moderately heritable, and genomewide association studies (GWAS) have identified risk genes in European, African, and Asian populations. The most reproducibly identified risk genes affect alcohol metabolism. Well-known functional variants at the gene encoding alcohol dehydrogenase B and other alcohol dehydrogenases affect risk in European and African ancestry populations. Similarly, variants mapped to these same genes and a well-known null variant that maps to the gene that encodes aldehyde dehydrogenase 2 (ALDH2) also affect risk in various Asian populations. In this study, we completed the first GWAS for 3 traits related to alcohol use in a Thai population recruited initially for studies of methamphetamine dependence. METHODS All subjects were evaluated with the Thai version of the Semi-Structured Assessment for Drug Dependence and Alcoholism (SSADDA). A total of 1,045 subjects were available for analysis. Three traits were analyzed: flushing, maximum number of alcoholic beverages consumed in any lifetime 24-hour period ("MAXDRINKS"), and DSM-IV alcohol dependence criterion count. We also conducted a pleiotropy analysis with major depression, the only other psychiatric trait where summary statistics from a large-scale Asian-population GWAS are available. RESULTS All 3 traits showed genomewide significant association with variants near ALDH2, with significance ranging from 2.01 × 10-14 (for flushing; lead single nucleotide polymorphism (SNP) PTPN11* rs143894582) to pmeta = 5.80 × 10-10 (for alcohol dependence criterion count; lead SNP rs149212747). These lead SNPs flank rs671 and span a region of over a megabase, illustrating the need for prior biological information in identifying the actual effect SNP, rs671. We also identified significant pleiotropy between major depression and flushing. CONCLUSIONS These results are consistent with prior findings in Asian populations and add new information regarding alcohol use-depression pleiotropy.
Collapse
Affiliation(s)
- Joel Gelernter
- Department of Psychiatry , Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry , VA Connecticut Healthcare System, West Haven, Connecticut.,Departments of Genetics and Neuroscience , Yale University School of Medicine, New Haven, Connecticut
| | - Hang Zhou
- Department of Psychiatry , Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry , VA Connecticut Healthcare System, West Haven, Connecticut
| | - Yaira Z Nuñez
- Department of Psychiatry , Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry , VA Connecticut Healthcare System, West Haven, Connecticut
| | - Apiwat Mutirangura
- Department of Anatomy , Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Robert T Malison
- Department of Psychiatry , Yale University School of Medicine, New Haven, Connecticut.,Clinical Neuroscience Research Unit , Connecticut Mental Health Center, New Haven, Connecticut
| | - Rasmon Kalayasiri
- Department of Psychiatry , King Chulalongkorn Memorial Hospital, Bangkok, Thailand.,Department of Psychiatry , Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW With the advent of the genome-wide association study (GWAS), our understanding of the genetics of addiction has made significant strides forward. Here, we summarize genetic loci containing variants identified at genome-wide statistical significance (P < 5 × 10-8) and independently replicated, review evidence of functional or regulatory effects for GWAS-identified variants, and outline multi-omics approaches to enhance discovery and characterize addiction loci. RECENT FINDINGS Replicable GWAS findings span 11 genetic loci for smoking, eight loci for alcohol, and two loci for illicit drugs combined and include missense functional variants and noncoding variants with regulatory effects in human brain tissues traditionally viewed as addiction-relevant (e.g., prefrontal cortex [PFC]) and, more recently, tissues often overlooked (e.g., cerebellum). GWAS analyses have discovered several novel, replicable variants contributing to addiction. Using larger sample sizes from harmonized datasets and new approaches to integrate GWAS with multiple 'omics data across human brain tissues holds great promise to significantly advance our understanding of the biology underlying addiction.
Collapse
Affiliation(s)
- Dana B Hancock
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Division, RTI International, 3040 East Cornwallis Road, P. O. Box 12194, Research Triangle Park, NC, 27709, USA.
| | - Christina A Markunas
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Division, RTI International, 3040 East Cornwallis Road, P. O. Box 12194, Research Triangle Park, NC, 27709, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Eric O Johnson
- Fellow Program and Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
42
|
Polimanti R, Gelernter J. ADH1B: From alcoholism, natural selection, and cancer to the human phenome. Am J Med Genet B Neuropsychiatr Genet 2018; 177:113-125. [PMID: 28349588 PMCID: PMC5617762 DOI: 10.1002/ajmg.b.32523] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/18/2022]
Abstract
The ADH1B (Alcohol Dehydrogenase 1B (class I), Beta Polypeptide) gene and its best-known functional alleles, Arg48His (rs1229984, ADH1B*2) and Arg370Cys (rs2066702, ADH1B*3), have been investigated in relation to many phenotypic traits; most frequently including alcohol metabolism and alcohol drinking behaviors, but also human evolution, liver function, cancer, and, recently, the comprehensive human phenome. To understand ADH1B functions and consequences, we provide here a bioinformatic analysis of its gene regulation and molecular functions, literature review of studies focused on this gene, and a discussion regarding future research perspectives. Certain ADH1B alleles have large effects on alcohol metabolism, and this relationship particularly encourages further investigations in relation to alcoholism and alcohol-associated cancer to understand better the mechanisms by which alcohol metabolism contributes to alcohol abuse and carcinogenesis. We also observed that ADH1B has complex mechanisms that regulate its expression across multiple human tissues, and these may be involved in cardiac and metabolic traits. Evolutionary data strongly suggest that the selection signatures at the ADH1B locus are primarily related to effects other than those on alcohol metabolism. This is also supported by the involvement of ADH1B in multiple molecular pathways and by the findings of our recent phenome-wide association study. Accordingly, future studies should also investigate other functions of ADH1B potentially relevant for the human phenome. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renato Polimanti
- Department of Psychiatry, Yale School of Medicine and VA CT Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine and VA CT Healthcare Center, West Haven, CT, USA
- Department of Genetics, Yale School of Medicine, West Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, West Haven, CT, USA
| |
Collapse
|
43
|
Prom-Wormley EC, Ebejer J, Dick DM, Bowers MS. The genetic epidemiology of substance use disorder: A review. Drug Alcohol Depend 2017; 180:241-259. [PMID: 28938182 PMCID: PMC5911369 DOI: 10.1016/j.drugalcdep.2017.06.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Substance use disorder (SUD) remains a significant public health issue. A greater understanding of how genes and environment interact to regulate phenotypes comprising SUD will facilitate directed treatments and prevention. METHODS The literature studying the neurobiological correlates of SUD with a focus on the genetic and environmental influences underlying these mechanisms was reviewed. Results from twin/family, human genetic association, gene-environment interaction, epigenetic literature, phenome-wide association studies are summarized for alcohol, nicotine, cannabinoids, cocaine, and opioids. RESULTS There are substantial genetic influences on SUD that are expected to influence multiple neurotransmission pathways, and these influences are particularly important within the dopaminergic system. Genetic influences involved in other aspects of SUD etiology including drug processing and metabolism are also identified. Studies of gene-environment interaction emphasize the importance of environmental context in SUD. Epigenetic studies indicate drug-specific changes in gene expression as well as differences in gene expression related to the use of multiple substances. Further, gene expression is expected to differ by stage of SUD such as substance initiation versus chronic substance use. While a substantial literature has developed for alcohol and nicotine use disorders, there is comparatively less information for other commonly abused substances. CONCLUSIONS A better understanding of genetically-mediated mechanisms involved in the neurobiology of SUD provides increased opportunity to develop behavioral and biologically based treatment and prevention of SUD.
Collapse
Affiliation(s)
- Elizabeth C Prom-Wormley
- Dvision of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, PO Box 980212, Richmond, VA 23298-0212, USA.
| | - Jane Ebejer
- School of Cognitive Behavioural and Social Sciences, University of New England, Armidale, NSW 2350, Australia
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, PO Box 842509, Richmond, VA 23284-2509, USA
| | - M Scott Bowers
- Faulk Center for Molecular Therapeutics, Biomedical Engeneering, Northwestern University, Evanston, IL 60201, USA
| |
Collapse
|
44
|
Kong X, Deng H, Gong S, Alston T, Kong Y, Wang J. Lack of associations of the opioid receptor mu 1 (OPRM1) A118G polymorphism (rs1799971) with alcohol dependence: review and meta-analysis of retrospective controlled studies. BMC MEDICAL GENETICS 2017; 18:120. [PMID: 29070014 PMCID: PMC5657079 DOI: 10.1186/s12881-017-0478-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022]
Abstract
Background Studies have sought associations of the opioid receptor mu 1 (OPRM1) A118G polymorphism (rs1799971) with alcohol-dependence, but findings are inconsistent. We summarize the information as to associations of rs1799971 (A > G) and the alcohol-dependence. Methods Systematically, we reviewed related literatures using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. Embase, PubMed, Web of Knowledge, and Chinese National Knowledge Infrastructure (CNKI) databases were searched using select medical subject heading (MeSH) terms to identify all researches focusing on the present topic up to September 2016. Odds ratios (ORs) along with the 95% confidence interval (95% CI) were estimated in allele model, homozygote model, heterozygote model, dominant model and recessive model. Ethnicity-specific subgroup-analysis, sensitivity analysis, heterogeneity description, and publication-bias assessment were also analyzed. Results There were 17 studies, including 9613 patients in the present meta-analysis. The ORs in the 5 genetic-models were 1.037 (95% CI: 0.890, 1.210; p = 0.64), 1.074 (95% CI: 0.831, 1.387; p = 0.586), 1.155 (95% CI: 0.935, 1.427; p = 0.181), 1.261 (95% CI: 1.008, 1.578; p = 0.042), 0.968 (95% CI: 0.758, 1.236; p = 0.793), respectively. An association is significant in the dominant model, but there is no statistical significance upon ethnicity-specific subgroup analysis. Conclusion The rs1799971 (A > G) is not strongly associated with alcohol-dependence. However, there are study heterogeneities and limited sample sizes.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, 55 Fruit Street, Boston, MA, 02114-3117, USA.,Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyangqu, Panjiayuan, Beijing, People's Republic of China
| | - Hao Deng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, 55 Fruit Street, Boston, MA, 02114-3117, USA
| | - Shun Gong
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, PLA Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 1249 Boylston St, Boston, MA, 02215, USA
| | - Theodore Alston
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, 55 Fruit Street, Boston, MA, 02114-3117, USA
| | - Yanguo Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China.
| | - Jingping Wang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, 55 Fruit Street, Boston, MA, 02114-3117, USA.
| |
Collapse
|
45
|
Yeung EW, Craggs JG, Gizer IR. Comorbidity of Alcohol Use Disorder and Chronic Pain: Genetic Influences on Brain Reward and Stress Systems. Alcohol Clin Exp Res 2017; 41:1831-1848. [PMID: 29048744 DOI: 10.1111/acer.13491] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/25/2017] [Indexed: 01/10/2023]
Abstract
Alcohol use disorder (AUD) is highly comorbid with chronic pain (CP). Evidence has suggested that neuroadaptive processes characterized by reward deficit and stress surfeit are involved in the development of AUD and pain chronification. Neurological data suggest that shared genetic architecture associated with the reward and stress systems may contribute to the comorbidity of AUD and CP. This monograph first delineates the prevailing theories of the development of AUD and pain chronification focusing on the reward and stress systems. It then provides a brief summary of relevant neurological findings followed by an evaluation of evidence documented by molecular genetic studies. Candidate gene association studies have provided some initial support for the genetic overlap between AUD and CP; however, these results must be interpreted with caution until studies with sufficient statistical power are conducted and replications obtained. Genomewide association studies have suggested a number of genes (e.g., TBX19, HTR7, and ADRA1A) that are either directly or indirectly related to the reward and stress systems in the AUD and CP literature. Evidence reviewed in this monograph suggests that shared genetic liability underlying the comorbidity between AUD and CP, if present, is likely to be complex. As the advancement in molecular genetic methods continues, future studies may show broader central nervous system involvement in AUD-CP comorbidity.
Collapse
Affiliation(s)
- Ellen W Yeung
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine, California
| | - Jason G Craggs
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,School of Health Professions, University of Missouri, Columbia, Missouri
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
46
|
Wolf JM, Simon D, Béria JU, Tietzmann DC, Stein AT, Lunge VR. Analysis of the Association of Nonsynonymous Polymorphisms in ADH Genes with Hazardous Drinking in HIV-1-Positive Individuals. Alcohol Clin Exp Res 2017; 41:1866-1874. [PMID: 28833276 DOI: 10.1111/acer.13486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/16/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Hazardous drinking (HD) is a serious health problem in people infected with human immunodeficiency virus type 1 (HIV-1). Single nucleotide polymorphisms (SNPs) in alcohol dehydrogenase (ADH) genes have been associated with HD in different populations, but there were no data about this in HIV-1-positive individuals. This study investigated the association of 4 nonsynonymous SNPs in ADH genes (Arg48His and Arg370Cys in ADH1B gene; Arg272Gln and Ile350Val in ADH1C gene) with HD in people living with HIV-1. METHODS This case-control study included 365 HIV-1-positive individuals (121 with HD and 244 without HD). Sociodemographic variables were collected with a standardized individual questionnaire. HD (score ≥8) and binge drinking (BD) (drinks on the same occasion ≥5) were detected with the Alcohol Use Disorders Identification Test. The 4 SNPs were genotyped by the polymerase chain reaction-restriction fragment length polymorphism method. Adjusted odds ratios (aORs) and 95% confidence intervals (CIs) were calculated using logistic regression analysis. The Bonferroni correction was used (considering the 4 SNPs studied). RESULTS There were no significant differences in the frequencies of Arg370Cys, Arg272Gln, and Ile350Val polymorphisms between HD cases and controls. Otherwise, Arg/His genotype (rs1229984) in ADH1B gene showed a protective effect against HD (aOR = 0.36; 95% CI: 0.14 to 0.90) and BD (aOR = 0.49; 95% CI: 0.21 to 0.95). Nevertheless, these differences were no longer significant after Bonferroni correction. CONCLUSIONS The results of this study suggest a possible effect of the Arg48His genotype on the protection against HD in HIV-1-positive individuals.
Collapse
Affiliation(s)
- Jonas Michel Wolf
- Laboratório de Diagnóstico Molecular , Universidade Luterana do Brasil (ULBRA), Canoas, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular Aplicada à Saúde , Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - Daniel Simon
- Programa de Pós-Graduação em Biologia Celular e Molecular Aplicada à Saúde , Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | | | | | - Airton Tetelbom Stein
- Fundação Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre, Brazil
| | - Vagner Ricardo Lunge
- Laboratório de Diagnóstico Molecular , Universidade Luterana do Brasil (ULBRA), Canoas, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular Aplicada à Saúde , Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| |
Collapse
|
47
|
Jorgenson E, Thai KK, Hoffmann TJ, Sakoda LC, Kvale MN, Banda Y, Schaefer C, Risch N, Mertens J, Weisner C, Choquet H. Genetic contributors to variation in alcohol consumption vary by race/ethnicity in a large multi-ethnic genome-wide association study. Mol Psychiatry 2017; 22:1359-1367. [PMID: 28485404 PMCID: PMC5568932 DOI: 10.1038/mp.2017.101] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/03/2017] [Accepted: 03/27/2017] [Indexed: 01/08/2023]
Abstract
Alcohol consumption is a complex trait determined by both genetic and environmental factors, and is correlated with the risk of alcohol use disorders. Although a small number of genetic loci have been reported to be associated with variation in alcohol consumption, genetic factors are estimated to explain about half of the variance in alcohol consumption, suggesting that additional loci remain to be discovered. We conducted a genome-wide association study (GWAS) of alcohol consumption in the large Genetic Epidemiology Research in Adult Health and Aging (GERA) cohort, in four race/ethnicity groups: non-Hispanic whites, Hispanic/Latinos, East Asians and African Americans. We examined two statistically independent phenotypes reflecting subjects' alcohol consumption during the past year, based on self-reported information: any alcohol intake (drinker/non-drinker status) and the regular quantity of drinks consumed per week (drinks/week) among drinkers. We assessed these two alcohol consumption phenotypes in each race/ethnicity group, and in a combined trans-ethnic meta-analysis comprising a total of 86 627 individuals. We observed the strongest association between the previously reported single nucleotide polymorphism (SNP) rs671 in ALDH2 and alcohol drinker status (odd ratio (OR)=0.40, P=2.28 × 10-72) in East Asians, and also an effect on drinks/week (beta=-0.17, P=5.42 × 10-4) in the same group. We also observed a genome-wide significant association in non-Hispanic whites between the previously reported SNP rs1229984 in ADH1B and both alcohol consumption phenotypes (OR=0.79, P=2.47 × 10-20 for drinker status and beta=-0.19, P=1.91 × 10-35 for drinks/week), which replicated in Hispanic/Latinos (OR=0.72, P=4.35 × 10-7 and beta=-0.21, P=2.58 × 10-6, respectively). Although prior studies reported effects of ADH1B and ALDH2 on lifetime measures, such as risk of alcohol dependence, our study adds further evidence of the effect of the same genes on a cross-sectional measure of average drinking. Our trans-ethnic meta-analysis confirmed recent findings implicating the KLB and GCKR loci in alcohol consumption, with strongest associations observed for rs7686419 (beta=-0.04, P=3.41 × 10-10 for drinks/week and OR=0.96, P=4.08 × 10-5 for drinker status), and rs4665985 (beta=0.04, P=2.26 × 10-8 for drinks/week and OR=1.04, P=5 × 10-4 for drinker status), respectively. Finally, we also obtained confirmatory results extending previous findings implicating AUTS2, SGOL1 and SERPINC1 genes in alcohol consumption traits in non-Hispanic whites.
Collapse
Affiliation(s)
- Eric Jorgenson
- Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Khanh K. Thai
- Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Thomas J. Hoffmann
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lori C. Sakoda
- Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Mark N. Kvale
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Yambazi Banda
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | | | - Neil Risch
- Kaiser Permanente Division of Research, Oakland, CA, USA,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | | | - Constance Weisner
- Kaiser Permanente Division of Research, Oakland, CA, USA,Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hélène Choquet
- Kaiser Permanente Division of Research, Oakland, CA, USA
| |
Collapse
|
48
|
Treutlein J, Frank J, Streit F, Reinbold CS, Juraeva D, Degenhardt F, Rietschel L, Witt SH, Forstner AJ, Ridinger M, Strohmaier J, Wodarz N, Dukal H, Foo JC, Hoffmann P, Herms S, Heilmann-Heimbach S, Soyka M, Maier W, Gaebel W, Dahmen N, Scherbaum N, Müller-Myhsok B, Lucae S, Ising M, Stickel F, Berg T, Roggenbuck U, Jöckel KH, Scholz H, Zimmermann US, Buch S, Sommer WH, Spanagel R, Brors B, Cichon S, Mann K, Kiefer F, Hampe J, Rosendahl J, Nöthen MM, Rietschel M. Genetic Contribution to Alcohol Dependence: Investigation of a Heterogeneous German Sample of Individuals with Alcohol Dependence, Chronic Alcoholic Pancreatitis, and Alcohol-Related Cirrhosis. Genes (Basel) 2017; 8:E183. [PMID: 28714907 PMCID: PMC5541316 DOI: 10.3390/genes8070183] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/11/2017] [Indexed: 01/14/2023] Open
Abstract
The present study investigated the genetic contribution to alcohol dependence (AD) using genome-wide association data from three German samples. These comprised patients with: (i) AD; (ii) chronic alcoholic pancreatitis (ACP); and (iii) alcohol-related liver cirrhosis (ALC). Single marker, gene-based, and pathway analyses were conducted. A significant association was detected for the ADH1B locus in a gene-based approach (puncorrected = 1.2 × 10-6; pcorrected = 0.020). This was driven by the AD subsample. No association with ADH1B was found in the combined ACP + ALC sample. On first inspection, this seems surprising, since ADH1B is a robustly replicated risk gene for AD and may therefore be expected to be associated also with subgroups of AD patients. The negative finding in the ACP + ALC sample, however, may reflect genetic stratification as well as random fluctuation of allele frequencies in the cases and controls, demonstrating the importance of large samples in which the phenotype is well assessed.
Collapse
Affiliation(s)
- Jens Treutlein
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Céline S Reinbold
- Human Genomics Research Group, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland.
| | - Dilafruz Juraeva
- Division of Applied Bioinformatics (G200), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
| | - Liz Rietschel
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3000 Bern 60, Switzerland.
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Andreas J Forstner
- Human Genomics Research Group, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
- Department of Psychiatry (U.P.K.), University of Basel, CH-4002 Basel, Switzerland.
| | - Monika Ridinger
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany.
- Psychiatric Health Care Aargau, 5210 Windisch, Switzerland.
| | - Jana Strohmaier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Norbert Wodarz
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany.
| | - Helene Dukal
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Jerome C Foo
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Per Hoffmann
- Human Genomics Research Group, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
| | - Stefan Herms
- Human Genomics Research Group, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
| | - Michael Soyka
- Private Hospital Meiringen, 3860 Meiringen, Switzerland.
- Psychiatric Hospital, Ludwig Maximilians University, 80336 Munich, Germany.
| | - Wolfgang Maier
- Department of Psychiatry, University of Bonn, 53105 Bonn, Germany.
| | - Wolfgang Gaebel
- Department of Psychiatry and Psychotherapy, University of Düsseldorf, 40629 Düsseldorf, Germany.
| | - Norbert Dahmen
- Department of Psychiatry, University of Mainz, 55131 Mainz, Germany.
| | - Norbert Scherbaum
- LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany.
| | - Bertram Müller-Myhsok
- Department of Statistical Genetics, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany.
| | - Susanne Lucae
- Department of Psychiatric Pharmacogenetics, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany.
| | - Marcus Ising
- Department of Molecular Psychology, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany.
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, 8091 Zurich, Switzerland.
- Hirslanden Private Hospital, 8032 Hirslanden Zürich, Switzerland.
| | - Thomas Berg
- Section of Hepatology, University Hospital Leipzig, 04103 Leipzig, Germany.
| | - Ulla Roggenbuck
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital of Essen, 45122 Essen, Germany.
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital of Essen, 45122 Essen, Germany.
| | - Henrike Scholz
- Department of Animal Physiology, University of Cologne, 50674 Cologne, Germany.
| | - Ulrich S Zimmermann
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Dresden Technical University, 01307 Dresden, Germany.
| | - Stephan Buch
- Medical Department 1, University Hospital Dresden, TU Dresden, 01307 Dresden, Germany.
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Benedikt Brors
- Division of Applied Bioinformatics (G200), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sven Cichon
- Human Genomics Research Group, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland.
| | - Karl Mann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, TU Dresden, 01307 Dresden, Germany.
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University Halle, 06120 Halle (Saale), Germany.
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany.
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| |
Collapse
|
49
|
Emerging roles for ncRNAs in alcohol use disorders. Alcohol 2017; 60:31-39. [PMID: 28438526 DOI: 10.1016/j.alcohol.2017.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
Chronic alcohol exposure produces widespread neuroadaptations and alterations in gene expression in human alcoholics and animal models. Technological advances in the past decade have increasingly highlighted the role of non-protein-coding RNAs (ncRNAs) in the regulation of gene expression and function. These recently characterized molecules were discovered to mediate diverse processes in the central nervous system, from normal development and physiology to regulation of disease, including alcoholism and other psychiatric disorders. This review will investigate the recent studies in human alcoholics and rodent models that have profiled different classes of ncRNAs and their dynamic alcohol-dependent regulation in brain.
Collapse
|
50
|
Rinker JA, Mulholland PJ. Promising pharmacogenetic targets for treating alcohol use disorder: evidence from preclinical models. Pharmacogenomics 2017; 18:555-570. [PMID: 28346058 DOI: 10.2217/pgs-2016-0193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inherited genetic variants contribute to risk factors for developing an alcohol use disorder, and polymorphisms may inform precision medicine strategies for treating alcohol addiction. Targeting genetic mutations linked to alcohol phenotypes has provided promising initial evidence for reducing relapse rates in alcoholics. Although successful in some studies, there are conflicting findings and the reports of adverse effects may ultimately limit their clinical utility, suggesting that novel pharmacogenetic targets are necessary to advance precision medicine approaches. Here, we describe promising novel genetic variants derived from preclinical models of alcohol consumption and dependence that may uncover disease mechanisms that drive uncontrolled drinking and identify novel pharmacogenetic targets that facilitate therapeutic intervention for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|