1
|
Clark SL, Hartwell EE, Choi DS, Krystal JH, Messing RO, Ferguson LB. Next-generation biomarkers for alcohol consumption and alcohol use disorder diagnosis, prognosis, and treatment: A critical review. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:5-24. [PMID: 39532676 PMCID: PMC11747793 DOI: 10.1111/acer.15476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
This critical review summarizes the current state of omics-based biomarkers in the alcohol research field. We first provide definitions and background information on alcohol and alcohol use disorder (AUD), biomarkers, and "omic" technologies. We next summarize using (1) genetic information as risk/prognostic biomarkers for the onset of alcohol-related problems and the progression from regular drinking to problematic drinking (including AUD), (2) epigenetic information as diagnostic biomarkers for AUD and risk biomarkers for alcohol consumption, (3) transcriptomic information as diagnostic biomarkers for AUD, risk biomarkers for alcohol consumption, and (4) metabolomic information as diagnostic biomarkers for AUD, risk biomarkers for alcohol consumption, and predictive biomarkers for response to acamprosate in subjects with AUD. In the final section, the clinical implications of the findings are discussed, and recommendations are made for future research.
Collapse
Affiliation(s)
- Shaunna L. Clark
- Department of Psychiatry & Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Emily E. Hartwell
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, USA
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, USA
| | - Laura B. Ferguson
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, USA
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
2
|
Li X, Liu J, Boreland AJ, Kapadia S, Zhang S, Stillitano AC, Abbo Y, Clark L, Lai D, Liu Y, Barr PB, Meyers JL, Kamarajan C, Kuang W, Agrawal A, Slesinger PA, Dick D, Salvatore J, Tischfield J, Duan J, Edenberg HJ, Kreimer A, Hart RP, Pang ZP. Polygenic risk for alcohol use disorder affects cellular responses to ethanol exposure in a human microglial cell model. SCIENCE ADVANCES 2024; 10:eado5820. [PMID: 39514655 PMCID: PMC11546823 DOI: 10.1126/sciadv.ado5820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Polygenic risk scores (PRSs) assess genetic susceptibility to alcohol use disorder (AUD), yet their molecular implications remain underexplored. Neuroimmune interactions, particularly in microglia, are recognized as notable contributors to AUD pathophysiology. We investigated the interplay between AUD PRS and ethanol in human microglia derived from iPSCs from individuals with AUD high-PRS (diagnosed with AUD) or low-PRS (unaffected). Ethanol exposure induced elevated CD68 expression and morphological changes in microglia, with differential responses between high-PRS and low-PRS microglial cells. Transcriptomic analysis revealed expression differences in MHCII complex and phagocytosis-related genes following ethanol exposure; high-PRS microglial cells displayed enhanced phagocytosis and increased CLEC7A expression, unlike low-PRS microglial cells. Synapse numbers in cocultures of induced neurons with microglia after alcohol exposure were lower in high-RPS cocultures, suggesting possible excess synapse pruning. This study provides insights into the intricate relationship between AUD PRS, ethanol, and microglial function, potentially influencing neuronal functions in developing AUD.
Collapse
Affiliation(s)
- Xindi Li
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Jiayi Liu
- Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Andrew J. Boreland
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Sneha Kapadia
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Alessandro C. Stillitano
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Yara Abbo
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Lorraine Clark
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Dongbing Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter B. Barr
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Chella Kamarajan
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Weipeng Kuang
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washinton University School of Medicine, Saint Louis, MO 63108, USA
| | - Paul A. Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Danielle Dick
- Department of Psychiatry, Rutgers University Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Jessica Salvatore
- Department of Psychiatry, Rutgers University Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Jay Tischfield
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Howard J. Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anat Kreimer
- Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ronald P. Hart
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
3
|
Gerring ZF, Thorp JG, Treur JL, Verweij KJH, Derks EM. The genetic landscape of substance use disorders. Mol Psychiatry 2024; 29:3694-3705. [PMID: 38811691 PMCID: PMC11541208 DOI: 10.1038/s41380-024-02547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 05/31/2024]
Abstract
Substance use disorders represent a significant public health concern with considerable socioeconomic implications worldwide. Twin and family-based studies have long established a heritable component underlying these disorders. In recent years, genome-wide association studies of large, broadly phenotyped samples have identified regions of the genome that harbour genetic risk variants associated with substance use disorders. These regions have enabled the discovery of putative causal genes and improved our understanding of genetic relationships among substance use disorders and other traits. Furthermore, the integration of these data with clinical information has yielded promising insights into how individuals respond to medications, allowing for the development of personalized treatment approaches based on an individual's genetic profile. This review article provides an overview of recent advances in the genetics of substance use disorders and demonstrates how genetic data may be used to reduce the burden of disease and improve public health outcomes.
Collapse
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics Laboratory, Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jackson G Thorp
- Translational Neurogenomics Laboratory, Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jorien L Treur
- Department of Psychiatry, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Karin J H Verweij
- Department of Psychiatry, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Eske M Derks
- Translational Neurogenomics Laboratory, Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Jennings MV, Martínez-Magaña JJ, Courchesne-Krak NS, Cupertino RB, Vilar-Ribó L, Bianchi SB, Hatoum AS, Atkinson EG, Giusti-Rodriguez P, Montalvo-Ortiz JL, Gelernter J, Artigas MS, Elson SL, Edenberg HJ, Fontanillas P, Palmer AA, Sanchez-Roige S. A phenome-wide association and Mendelian randomisation study of alcohol use variants in a diverse cohort comprising over 3 million individuals. EBioMedicine 2024; 103:105086. [PMID: 38580523 PMCID: PMC11121167 DOI: 10.1016/j.ebiom.2024.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Alcohol consumption is associated with numerous negative social and health outcomes. These associations may be direct consequences of drinking, or they may reflect common genetic factors that influence both alcohol consumption and other outcomes. METHODS We performed exploratory phenome-wide association studies (PheWAS) of three of the best studied protective single nucleotide polymorphisms (SNPs) in genes encoding ethanol metabolising enzymes (ADH1B: rs1229984-T, rs2066702-A; ADH1C: rs698-T) using up to 1109 health outcomes across 28 phenotypic categories (e.g., substance-use, mental health, sleep, immune, cardiovascular, metabolic) from a diverse 23andMe cohort, including European (N ≤ 2,619,939), Latin American (N ≤ 446,646) and African American (N ≤ 146,776) populations to uncover new and perhaps unexpected associations. These SNPs have been consistently implicated by both candidate gene studies and genome-wide association studies of alcohol-related behaviours but have not been investigated in detail for other relevant phenotypes in a hypothesis-free approach in such a large cohort of multiple ancestries. To provide insight into potential causal effects of alcohol consumption on the outcomes significant in the PheWAS, we performed univariable two-sample and one-sample Mendelian randomisation (MR) analyses. FINDINGS The minor allele rs1229984-T, which is protective against alcohol behaviours, showed the highest number of PheWAS associations across the three cohorts (N = 232, European; N = 29, Latin American; N = 7, African American). rs1229984-T influenced multiple domains of health. We replicated associations with alcohol-related behaviours, mental and sleep conditions, and cardio-metabolic health. We also found associations with understudied traits related to neurological (migraines, epilepsy), immune (allergies), musculoskeletal (fibromyalgia), and reproductive health (preeclampsia). MR analyses identified evidence of causal effects of alcohol consumption on liability for 35 of these outcomes in the European cohort. INTERPRETATION Our work demonstrates that polymorphisms in genes encoding alcohol metabolising enzymes affect multiple domains of health beyond alcohol-related behaviours. Understanding the underlying mechanisms of these effects could have implications for treatments and preventative medicine. FUNDING MVJ, NCK, SBB, SSR and AAP were supported by T32IR5226 and 28IR-0070. SSR was also supported by NIDA DP1DA054394. NCK and RBC were also supported by R25MH081482. ASH was supported by funds from NIAAA K01AA030083. JLMO was supported by VA 1IK2CX002095. JLMO and JJMM were also supported by NIDA R21DA050160. JJMM was also supported by the Kavli Postdoctoral Award for Academic Diversity. EGA was supported by K01MH121659 from the NIMH/NIH, the Caroline Wiess Law Fund for Research in Molecular Medicine and the ARCO Foundation Young Teacher-Investigator Fund at Baylor College of Medicine. MSA was supported by the Instituto de Salud Carlos III and co-funded by the European Union Found: Fondo Social Europeo Plus (FSE+) (P19/01224, PI22/00464 and CP22/00128).
Collapse
Affiliation(s)
- Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - José Jaime Martínez-Magaña
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, Orange, West Haven, CT, USA
| | | | - Renata B Cupertino
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Laura Vilar-Ribó
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Sevim B Bianchi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Hatoum
- Department of Psychology & Brain Sciences, Washington University in St. Louis, St Louis, MO, USA
| | - Elizabeth G Atkinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Paola Giusti-Rodriguez
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA
| | - Janitza L Montalvo-Ortiz
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, Orange, West Haven, CT, USA; National Center of Posttraumatic Stress Disorder, VA CT Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- VA CT Healthcare Center, Department Psychiatry, West Haven, CT, USA; Departments Psychiatry, Genetics, and Neuroscience, Yale Univ. School of Medicine, New Haven, CT, USA
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain; Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | | | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
5
|
Clapp Sullivan ML, Schwaba T, Harden KP, Grotzinger AD, Nivard MG, Tucker-Drob EM. Beyond the factor indeterminacy problem using genome-wide association data. Nat Hum Behav 2024; 8:205-218. [PMID: 38225407 PMCID: PMC10922726 DOI: 10.1038/s41562-023-01789-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/20/2023] [Indexed: 01/17/2024]
Abstract
Latent factors, such as general intelligence, depression and risk tolerance, are invoked in nearly all social science research where a construct is measured via aggregation of symptoms, question responses or other measurements. Because latent factors cannot be directly observed, they are inferred by fitting a specific model to empirical patterns of correlations among measured variables. A long-standing critique of latent factor theories is that the correlations used to infer latent factors can be produced by alternative data-generating mechanisms that do not include latent factors. This is referred to as the factor indeterminacy problem. Researchers have recently begun to overcome this problem by using information on the associations between individual genetic variants and measured variables. We review historical work on the factor indeterminacy problem and describe recent efforts in genomics to rigorously test the validity of latent factors, advancing the understanding of behavioural science constructs.
Collapse
Affiliation(s)
| | - Ted Schwaba
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Andrew D Grotzinger
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado at Boulder, Boulder, CO, USA
| | - Michel G Nivard
- Department of Biological Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Elliot M Tucker-Drob
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Hill SY, Hostyk J. A whole exome sequencing study to identify rare variants in multiplex families with alcohol use disorder. Front Psychiatry 2023; 14:1216493. [PMID: 37915799 PMCID: PMC10616827 DOI: 10.3389/fpsyt.2023.1216493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
Background Alcohol use disorder (AUD) runs in families and is accompanied by genetic variation. Some families exhibit an extreme susceptibility in which multiple cases are found and often with an early onset of the disorder. Large scale genome-wide association studies have identified several genes with impressive statistical probabilities. Most of these genes are common variants. Our goal was to perform exome sequencing in families characterized by multiple cases (multiplex families) to determine if rare variants might be segregating with disease status. Methods A case-control approach was used to leverage the power of a large control sample of unrelated individuals (N = 8,983) with exome sequencing [Institute for Genomic Medicine (IGM)], for comparison with probands with AUD (N = 53) from families selected for AUD multiplex status. The probands were sequenced at IGM using similar protocols to those used for the archival controls. Specifically, the presence of a same-sex pair of adult siblings with AUD was the minimal criteria for inclusion. Using a gene-based collapsing analysis strategy, a search for qualifying variants within the sequence data was undertaken to identify ultra-rare non-synonymous variants. Results We searched 18,666 protein coding genes to identify an excess of rare deleterious genetic variation using whole exome sequence data in the 53 AUD individuals from a total of 282 family members. To complete a case/control analysis of unrelated individuals, probands were compared to unrelated controls. Case enrichment for 16 genes with significance at 10-4 and one at 10-5 are plausible candidates for follow-up studies. Six genes were ultra rare [minor allele frequency (MAF) of 0.0005]: CDSN, CHRNA9, IFT43, TLR6, SELENBP1, and GMPPB. Eight genes with MAF of 0.001: ZNF514, OXGR1, DIEXF, TMX4, MTBP, PON2, CRHBP, and ANKRD46 were identified along with three protein-truncating variants associated with loss-of-function: AGTRAP, ANKRD46, and PPA1. Using an ancestry filtered control group (N = 2,814), nine genes were found; three were also significant in the comparison to the larger control group including CHRNA9 previously implicated in alcohol and nicotine dependence. Conclusion This study implicates ultra-rare loss-of-function genes in AUD cases. Among the genes identified include those previously reported for nicotine and alcohol dependence (CHRNA9 and CRHBP).
Collapse
Affiliation(s)
- Shirley Y. Hill
- Department of Psychiatry, Psychology and Human Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph Hostyk
- Institute for Genomic Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
7
|
Thomas NS, Salvatore JE, Kuo SIC, Aliev F, McCutcheon VV, Meyers JM, Bucholz KK, Brislin SJ, Chan G, Edenberg HJ, Kamarajan C, Kramer JR, Kuperman S, Pandey G, Plawecki MH, Schuckit MA, Dick DM. Genetic nurture effects for alcohol use disorder. Mol Psychiatry 2023; 28:759-766. [PMID: 36253439 PMCID: PMC10079179 DOI: 10.1038/s41380-022-01816-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022]
Abstract
We tested whether aspects of the childhood/adolescent home environment mediate genetic risk for alcohol problems within families across generations. Parental relationship discord and parental divorce were the focal environments examined. The sample included participants of European ancestry (N = 4806, 51% female) and African ancestry (N = 1960, 52% female) from the high-risk Collaborative Study on the Genetics of Alcoholism. Alcohol outcomes in the child generation included lifetime criterion counts for DSM-5 Alcohol Use Disorder (AUD), lifetime maximum drinks in 24 h, age at initiation of regular drinking, and age at first alcohol intoxication. Predictors in the parent generation included relationship discord, divorce, alcohol measures parallel to those in the child generation, and polygenic scores for alcohol problems. Parental polygenic scores were partitioned into alleles that were transmitted and non-transmitted to the child. The results from structural equation models were consistent with genetic nurture effects in European ancestry families. Exposure to parental relationship discord and parental divorce mediated, in part, the transmission of genetic risk for alcohol problems from parents to children to predict earlier ages regular drinking (βindirect = -0.018 [-0.026, -0.011]) and intoxication (βindirect = -0.015 [-0.023, -0.008]), greater lifetime maximum drinks (βindirect = 0.006 [0.002, 0.01]) and more lifetime AUD criteria (βindirect = 0.011 [0.006, 0.016]). In contrast, there was no evidence that parental alleles had indirect effects on offspring alcohol outcomes via parental relationship discord or divorce in the smaller number of families of African ancestry. In conclusion, parents transmit genetic risk for alcohol problems to their children not only directly, but also indirectly via genetically influenced aspects of the home environment. Further investigation of genetic nurture in non-European samples is needed.
Collapse
Affiliation(s)
- Nathaniel S Thomas
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.
| | - Jessica E Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA.
| | - Sally I-Chun Kuo
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Fazil Aliev
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Vivia V McCutcheon
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacquelyn M Meyers
- Department of Psychiatry and Behavioral Sciences, State University of New York Health Sciences University, Brooklyn, NY, USA
| | - Kathleen K Bucholz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah J Brislin
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Grace Chan
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Chella Kamarajan
- Department of Psychiatry and Behavioral Sciences, State University of New York Health Sciences University, Brooklyn, NY, USA
| | - John R Kramer
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Samuel Kuperman
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Gayathri Pandey
- Department of Psychiatry and Behavioral Sciences, State University of New York Health Sciences University, Brooklyn, NY, USA
| | | | - Marc A Schuckit
- Department of Psychiatry, University of California San Diego Medical School, San Diego, CA, USA
| | - Danielle M Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
8
|
Savage JE, Peterson RE, Aliev F, Dick DM. Genetic and environmental etiology of drinking motives in college students. Alcohol Clin Exp Res 2022; 46:1783-1796. [PMID: 36256465 PMCID: PMC9828131 DOI: 10.1111/acer.14930] [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: 04/04/2022] [Revised: 07/16/2022] [Accepted: 08/18/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Drinking motives are robust proximal predictors of alcohol use behaviors and may mediate distinct etiological pathways in the development of alcohol misuse. However, little is known about the genetic and environmental etiology of drinking motives themselves and their potential utility as endophenotypes. METHODS Here, we leverage a longitudinal study of college students from diverse racial/ethnic backgrounds (phenotypic N = 9889, genotypic N = 4855) to investigate the temporal stability and demographic and environmental predictors of four types of drinking motives (enhancement, social, coping, and conformity). Using genome-wide association study (GWAS) and in silico tools, we characterize their associated genes and genetic variants (single nucleotide polymorphisms or SNPs). RESULTS Drinking motives were stable across four years of college (ICC >0.74). Some robust environmental predictors of alcohol misuse (parental autonomy granting and peer deviance) were broadly associated with multiple types of drinking motives, while others (e.g., trauma exposure) were type specific. Genome-wide analyses indicated modest SNP-based heritability (14-22%, n.s.) and several suggestive genomic loci that corroborate findings from previous molecular genetic studies (e.g., PECR and SIRT4 genes), indicating possible differences in the genetic etiology of positive versus negative reinforcement drinking motives that align with an internalizing/externalizing typology of alcohol misuse. Coping motives were significantly genetically correlated with alcohol use disorder diagnoses (rg = 0.71, p = 0.001). However, results from the genetic analyses were largely underpowered to detect significant associations. CONCLUSIONS Drinking motives show promise as endophenotypes but require further investigation in larger samples to further our understanding of the etiology of alcohol misuse.
Collapse
Affiliation(s)
- Jeanne E. Savage
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA,Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive ResearchVrije UniversiteitAmsterdamNetherlands
| | - Roseann E. Peterson
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Fazil Aliev
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA,Faculty of BusinessKarabuk UniversityKarabukTurkey,Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers – The State University of New JerseyPiscatawayNew JerseyUSA
| | | | - Danielle M. Dick
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA,Department of Psychiatry, Robert Wood Johnson Medical SchoolRutgers – The State University of New JerseyPiscatawayNew JerseyUSA,Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA,College Behavioral and Emotional Health InstituteVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
9
|
Bajaj JS, Nagy LE. Natural History of Alcohol-Associated Liver Disease: Understanding the Changing Landscape of Pathophysiology and Patient Care. Gastroenterology 2022; 163:840-851. [PMID: 35598629 PMCID: PMC9509416 DOI: 10.1053/j.gastro.2022.05.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 12/02/2022]
Abstract
Alcohol use and consequent liver disease are major burdens that have worsened during the COVID-19 pandemic. There are several facets to the pathophysiology and clinical consequences of alcohol-use disorder (AUD) and progression to alcohol-associated liver disease (ALD) that require a concerted effort by clinicians and translational and basic science investigators. Several recent advances from bedside to bench and bench to bedside have been made in ALD. We focused this review on a case-based approach that provides a human context to these important advances across the spectrum of ALD.
Collapse
Affiliation(s)
- Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, Virginia; Central Virginia Veterans Healthcare System, Richmond, Virginia.
| | - Laura E Nagy
- Center for Liver Disease Research, Departments of Inflammation and Immunity and Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio; Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
10
|
Zhou H, Kalayasiri R, Sun Y, Nuñez YZ, Deng HW, Chen XD, Justice AC, Kranzler HR, Chang S, Lu L, Shi J, Sanichwankul K, Mutirangura A, Malison RT, Gelernter J. Genome-wide meta-analysis of alcohol use disorder in East Asians. Neuropsychopharmacology 2022; 47:1791-1797. [PMID: 35094024 PMCID: PMC9372033 DOI: 10.1038/s41386-022-01265-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022]
Abstract
Alcohol use disorder (AUD) is a leading cause of death and disability worldwide. Genome-wide association studies (GWAS) have identified ~30 AUD risk genes in European populations, but many fewer in East Asians. We conducted GWAS and genome-wide meta-analysis of AUD in 13,551 subjects with East Asian ancestry, using published summary data and newly genotyped data from five cohorts: (1) electronic health record (EHR)-diagnosed AUD in the Million Veteran Program (MVP) sample; (2) DSM-IV diagnosed alcohol dependence (AD) in a Han Chinese-GSA (array) cohort; (3) AD in a Han Chinese-Cyto (array) cohort; and (4) two AD Thai cohorts. The MVP and Thai samples included newly genotyped subjects from ongoing recruitment. In total, 2254 cases and 11,297 controls were analyzed. An AUD polygenic risk score was analyzed in an independent sample with 4464 East Asians (Genetic Epidemiology Research in Adult Health and Aging (GERA)). Phenotypes from survey data and ICD-9-CM diagnoses were tested for association with the AUD PRS. Two risk loci were detected: the well-known functional variant rs1229984 in ADH1B and rs3782886 in BRAP (near the ALDH2 gene locus) are the lead variants. AUD PRS was significantly associated with days per week of alcohol consumption (beta = 0.43, SE = 0.067, p = 2.47 × 10-10) and nominally associated with pack years of smoking (beta = 0.09, SE = 0.05, p = 4.52 × 10-2) and ever vs. never smoking (beta = 0.06, SE = 0.02, p = 1.14 × 10-2). This is the largest GWAS of AUD in East Asians to date. Building on previous findings, we were able to analyze pleiotropy, but did not identify any new risk regions, underscoring the importance of recruiting additional East Asian subjects for alcohol GWAS.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Rasmon Kalayasiri
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yan Sun
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yaira Z Nuñez
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Hong-Wen Deng
- Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Xiang-Ding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Amy C Justice
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Interdisciplinary Research on AIDS, Yale School of Public Health, New Haven, CT, USA
| | - Henry R Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China
| | | | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Robert T Malison
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA.
- Departments of Genetics and Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Zindler T, Frieling H, Fliedner L, Veer IM, Neyazi A, Awasthi S, Ripke S, Walter H, Friedel E. How alcohol makes the epigenetic clock tick faster and the clock reversing effect of abstinence. Addict Biol 2022; 27:e13198. [DOI: 10.1111/adb.13198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/22/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Tristan Zindler
- Department of Psychiatry, Social Psychiatry and Psychotherapy Hannover Medical School Hannover Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy Hannover Medical School Hannover Germany
| | - Lena Fliedner
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
| | - Ilya M. Veer
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
| | - Alexandra Neyazi
- Department of Psychiatry, Social Psychiatry and Psychotherapy Hannover Medical School Hannover Germany
| | - Swapnil Awasthi
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
| | - Stephan Ripke
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
| | - Henrik Walter
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
| | - Eva Friedel
- Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health, Charité Campus Mitte (CCM) Berlin Germany
- Berlin Institute of Health (BIH) Berlin Germany
| |
Collapse
|
12
|
Parker CC, Philip VM, Gatti DM, Kasparek S, Kreuzman AM, Kuffler L, Mansky B, Masneuf S, Sharif K, Sluys E, Taterra D, Taylor WM, Thomas M, Polesskaya O, Palmer AA, Holmes A, Chesler EJ. Genome-wide association mapping of ethanol sensitivity in the Diversity Outbred mouse population. Alcohol Clin Exp Res 2022; 46:941-960. [PMID: 35383961 DOI: 10.1111/acer.14825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/04/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND A strong predictor for the development of alcohol use disorder (AUD) is altered sensitivity to the intoxicating effects of alcohol. Individual differences in the initial sensitivity to alcohol are controlled in part by genetic factors. Mice offer a powerful tool to elucidate the genetic basis of behavioral and physiological traits relevant to AUD, but conventional experimental crosses have only been able to identify large chromosomal regions rather than specific genes. Genetically diverse, highly recombinant mouse populations make it possible to observe a wider range of phenotypic variation, offer greater mapping precision, and thus increase the potential for efficient gene identification. METHODS We have taken advantage of the Diversity Outbred (DO) mouse population to identify and precisely map quantitative trait loci (QTL) associated with ethanol sensitivity. We phenotyped 798 male J:DO mice for three measures of ethanol sensitivity: ataxia, hypothermia, and loss of the righting response. We used high-density MegaMUGA and GigaMUGA to obtain genotypes ranging from 77,808 to 143,259 SNPs. We also performed RNA sequencing in striatum to map expression QTLs and identify gene expression-trait correlations. We then applied a systems genetic strategy to identify narrow QTLs and construct the network of correlations that exists between DNA sequence, gene expression values, and ethanol-related phenotypes to prioritize our list of positional candidate genes. RESULTS We observed large amounts of phenotypic variation with the DO population and identified suggestive and significant QTLs associated with ethanol sensitivity on chromosomes 1, 2, and 16. The implicated regions were narrow (4.5-6.9 Mb in size) and each QTL explained ~4-5% of the variance. CONCLUSIONS Our results can be used to identify alleles that contribute to AUD in humans, elucidate causative biological mechanisms, or assist in the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Clarissa C Parker
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Vivek M Philip
- Center for Computational Sciences, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Daniel M Gatti
- Center for Computational Sciences, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Steven Kasparek
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Andrew M Kreuzman
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Lauren Kuffler
- Center for Mammalian Genetics, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Benjamin Mansky
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Sophie Masneuf
- Laboratory of Behavioral and Genomic Neuroscience, NIAAA, NIH, Rockville, MD, USA
| | - Kayvon Sharif
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Erica Sluys
- Laboratory of Behavioral and Genomic Neuroscience, NIAAA, NIH, Rockville, MD, USA
| | - Dominik Taterra
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Walter M Taylor
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Mary Thomas
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, Vermont, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, NIAAA, NIH, Rockville, MD, USA
| | - Elissa J Chesler
- Center for Mammalian Genetics, The Jackson Laboratory, Bar Harbor, Maine, USA
| |
Collapse
|
13
|
Maccioni P, Bratzu J, Lobina C, Acciaro C, Corrias G, Capra A, Carai MAM, Agabio R, Muntoni AL, Gessa GL, Colombo G. Exposure to an enriched environment reduces alcohol self-administration in Sardinian alcohol-preferring rats. Physiol Behav 2022; 249:113771. [PMID: 35247441 DOI: 10.1016/j.physbeh.2022.113771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 10/19/2022]
Abstract
Living in an enriched environment (EE) produces a notable impact on several rodent behaviors, including those motivated by drugs of abuse. This picture is somewhat less clear when referring to alcohol-motivated behaviors. With the intent of contributing to this research field with data from one of the few rat lines selectively bred for excessive alcohol consumption, the present study investigated the effect of EE on operant oral alcohol self-administration in Sardinian alcohol-preferring (sP) rats. Starting from Postnatal Day (PND) 21, male sP rats were kept under 3 different housing conditions: impoverished environment (IE; single housing in shoebox-like cages with no environmental enrichment); standard environment (SE; small colony cages with 3 rats and no environmental enrichment); EE (large colony cages with 6 rats and multiple elements of environmental enrichment, including 2 floors, ladders, maze, running wheels, and shelter). From PND 60, rats were exposed to different phases of shaping and training of alcohol self-administration. IE, SE, and EE rats were then compared under (i) fixed ratio (FR) 4 (FR4) schedule of alcohol reinforcement for 20 daily sessions and (ii) progressive ratio (PR) schedule of alcohol reinforcement in a final single session. Acquisition of the lever-responding task (shaping) was slower in EE than IE and SE rats, as the likely consequence of a "devaluation" of the novel stimuli provided by the operant chamber in comparison to those to which EE rats were continuously exposed in their homecage or an alteration, induced by EE, of the rat "emotionality" state when facing the novel environment represented by the operant chamber. Training of alcohol self-administration was slower in EE than IE rats, with SE rats displaying intermediate values. A similar ranking order (IE>SE>EE) was also observed in number of lever-responses for alcohol, amount of self-administered alcohol, and breakpoint for alcohol under FR4 and PR schedules of reinforcement. These data suggest that living in a complex environment reduced the reinforcing and motivational properties of alcohol in sP rats. These results are interpreted in terms of the reinforcing and motivational properties of the main components of EE (i.e., social interactions, physical activities, exploration, novelty) substituting, at least partially, for those of alcohol.
Collapse
Affiliation(s)
- Paola Maccioni
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy
| | - Jessica Bratzu
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy
| | - Carla Lobina
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy
| | - Carla Acciaro
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy
| | - Gianluigi Corrias
- Department of Physics, University of Cagliari, I-09042 Monserrato (CA), Italy
| | - Alessandro Capra
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, I-09042 Monserrato (CA), Italy
| | - Mauro A M Carai
- Cagliari Pharmacological Research, I-09127 Cagliari (CA), Italy
| | - Roberta Agabio
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, I-09042 Monserrato (CA), Italy
| | - Anna Lisa Muntoni
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy
| | - Gian Luigi Gessa
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy; Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, I-09042 Monserrato (CA), Italy
| | - Giancarlo Colombo
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, I-09042 Monserrato (CA), Italy.
| |
Collapse
|
14
|
Pastana LF, Silva TA, Gellen LPA, Vieira GM, de Assunção LA, Leitão LPC, da Silva NM, Coelho RDCC, de Alcântara AL, Vinagre LWMS, Rodrigues JCG, Borges Leal DFDV, Fernandes MR, de Souza SJ, Kroll JE, Ribeiro-dos-Santos AM, Burbano RMR, Guerreiro JF, de Assumpção PP, Ribeiro-dos-Santos ÂC, dos Santos SEB, dos Santos NPC. The Genomic Profile Associated with Risk of Severe Forms of COVID-19 in Amazonian Native American Populations. J Pers Med 2022; 12:jpm12040554. [PMID: 35455670 PMCID: PMC9027999 DOI: 10.3390/jpm12040554] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Genetic factors associated with COVID-19 disease outcomes are poorly understood. This study aimed to associate genetic variants in the SLC6A20, LZTFL1, CCR9, FYCO1, CXCR6, XCR1, and ABO genes with the risk of severe forms of COVID-19 in Amazonian Native Americans, and to compare the frequencies with continental populations. The study population was composed of 64 Amerindians from the Amazon region of northern Brazil. The difference in frequencies between the populations was analyzed using Fisher’s exact test, and the results were significant when p ≤ 0.05. We investigated 64 polymorphisms in 7 genes; we studied 47 genetic variants that were new or had impact predictions of high, moderate, or modifier. We identified 15 polymorphisms with moderate impact prediction in 4 genes (ABO, CXCR6, FYCO1, and SLC6A20). Among the variants analyzed, 18 showed significant differences in allele frequency in the NAM population when compared to others. We reported two new genetic variants with modifier impact in the Amazonian population that could be studied to validate the possible associations with COVID-19 outcomes. The genomic profile of Amazonian Native Americans may be associated with protection from severe forms of COVID-19. This work provides genomic data that may help forthcoming studies to improve COVID-19 outcomes.
Collapse
Affiliation(s)
- Lucas Favacho Pastana
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Thays Amâncio Silva
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Laura Patrícia Albarello Gellen
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Giovana Miranda Vieira
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Letícia Almeida de Assunção
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Luciana Pereira Colares Leitão
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Natasha Monte da Silva
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Rita de Cássia Calderaro Coelho
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Angélica Leite de Alcântara
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Lui Wallacy Morikawa Souza Vinagre
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Juliana Carla Gomes Rodrigues
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Diana Feio da Veiga Borges Leal
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Marianne Rodrigues Fernandes
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Sandro José de Souza
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal 59076-550, Brazil; (S.J.d.S.); (J.E.K.)
- BioME, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil
- Institute of Systems Genetics, West China Hospital, University of Sichuan, Chengdu 610041, China
| | - José Eduardo Kroll
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal 59076-550, Brazil; (S.J.d.S.); (J.E.K.)
| | - André Mauricio Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Rommel Mario Rodríguez Burbano
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - João Farias Guerreiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Paulo Pimentel de Assumpção
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Ândrea Campos Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Brazil; (A.M.R.-d.-S.); (J.F.G.); (Â.C.R.-d.-S.)
| | - Sidney Emanuel Batista dos Santos
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
| | - Ney Pereira Carneiro dos Santos
- Laboratório do Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil; (L.F.P.); (T.A.S.); (L.P.A.G.); (G.M.V.); (L.A.d.A.); (L.P.C.L.); (N.M.d.S.); (R.d.C.C.C.); (A.L.d.A.); (L.W.M.S.V.); (J.C.G.R.); (D.F.d.V.B.L.); (M.R.F.); (R.M.R.B.); (P.P.d.A.); (S.E.B.d.S.)
- Correspondence: ; Tel.: +55-(91)-98107-0850
| |
Collapse
|
15
|
Sakala K, Kasearu K, Katus U, Veidebaum T, Harro J. Association between platelet MAO activity and lifetime drug use in a longitudinal birth cohort study. Psychopharmacology (Berl) 2022; 239:327-337. [PMID: 35001146 DOI: 10.1007/s00213-021-06035-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE Platelet monoamine oxidase (MAO) activity, a marker of central serotonergic capacity, has been associated with a variety of problem behaviours. However, studies on platelet MAO activity and addictive drugs have not consistently linked MAO activity with addiction or reported to predict illicit substance use initiation or frequency. OBJECTIVES Platelet MAO activity and illicit drug use was examined in a longitudinal birth cohort study. METHODS The sample included both birth cohorts (original n = 1238) of the Estonian Children Personality Behaviour and Health Study. Longitudinal association from age 15 to 25 years between platelet MAO activity and lifetime drug use was analysed by mixed-effects regression models. Differences at ages 15, 18 and 25 were analysed by t-test. Cox proportional hazard regression analysis was used to assess the association between platelet MAO activity and the age of drug use initiation. RESULTS Male subjects who reported at least one drug use event had lower platelet MAO activity compared to nonusers, both in cross-sectional and longitudinal analyses. Males with low platelet MAO activity had started to use drugs at a younger age. Moreover, in male subjects who had experimented with illicit drugs only once in lifetime, low platelet MAO activity was also associated with higher risk at a younger age. In females, platelet MAO activity was not associated with drug use. CONCLUSION In males, low platelet MAO activity is associated with drug abuse primarily owing to risk-taking at early age.
Collapse
Affiliation(s)
- Katre Sakala
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia.,Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.,School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia
| | - Kairi Kasearu
- Institute of Social Studies, Faculty of Social Sciences, University of Tartu, Lossi 36, 51003, Tartu, Estonia
| | - Urmeli Katus
- Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Toomas Veidebaum
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia. .,Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14a, 50411, Tartu, Estonia.
| |
Collapse
|
16
|
The Vista of Application of Specific Anaphylaxis Accurate Diagnosis Based on DNA Single-Nucleotide Methylation Sites. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:8202068. [PMID: 34908915 PMCID: PMC8635942 DOI: 10.1155/2021/8202068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/21/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022]
Abstract
Anaphylaxis has rapidly spread around the world in the last several decades. Environmental factors seem to play a major role, and epigenetic marks, especially DNA methylation, get more attention. We discussed several GEO opening data classifications with TOP 100 specific methylation region values (normalized M-values on line) by machine learning, which are remarkable to classify specific anaphylaxis after monoallergen exposure. Then, we sequenced the whole-genome DNA methylation of six people (3 wormwood monoallergen atopic rhinitis patients and 3 normal-immune people) during the pollen season and analyzed the difference of the single nucleotide and DNA region. The results' divergences were obvious (the differential single nucleotides were mostly distributed in nongene regions but the differential DNA regions of GWAS, on the other hand), which may have caused most single nucleotides to be concealed in the regions' sequences. Therefore, we suggest that we should conduct more “pragmatic” and directly find special single-nucleotide changes after exposure to atopic allergens instead of complex correlativity. It is possible to try to use DNA methylation marks to accurately diagnose anaphylaxis and form a machine learning classification based on the single methylated CpGs.
Collapse
|
17
|
Vornholt E, Drake J, Mamdani M, McMichael G, Taylor ZN, Bacanu S, Miles MF, Vladimirov VI. Identifying a novel biological mechanism for alcohol addiction associated with circRNA networks acting as potential miRNA sponges. Addict Biol 2021; 26:e13071. [PMID: 34164896 PMCID: PMC8590811 DOI: 10.1111/adb.13071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/21/2021] [Accepted: 05/31/2021] [Indexed: 12/11/2022]
Abstract
Our lab and others have shown that chronic alcohol use leads to gene and miRNA expression changes across the mesocorticolimbic (MCL) system. Circular RNAs (circRNAs) are noncoding RNAs that form closed-loop structures and are reported to alter gene expression through miRNA sequestration, thus providing a potentially novel neurobiological mechanism for the development of alcohol dependence (AD). Genome-wide expression of circRNA was assessed in the nucleus accumbens (NAc) from 32 AD-matched cases/controls. Significant circRNAs (unadj. p ≤ 0.05) were identified via regression and clustered in circRNA networks via weighted gene co-expression network analysis (WGCNA). CircRNA interactions with previously generated mRNA and miRNA were detected via correlation and bioinformatic analyses. Significant circRNAs (N = 542) clustered in nine significant AD modules (FWER p ≤ 0.05), within which we identified 137 circRNA hubs. We detected 23 significant circRNA-miRNA-mRNA interactions (FDR ≤ 0.10). Among these, circRNA-406742 and miR-1200 significantly interact with the highest number of mRNA, including genes associated with neuronal functioning and alcohol addiction (HRAS, PRKCB, HOMER1, and PCLO). Finally, we integrate genotypic information that revealed 96 significant circRNA expression quantitative trait loci (eQTLs) (unadj. p ≤ 0.002) that showed significant enrichment within recent alcohol use disorder (AUD) and smoking genome-wide association study (GWAS). To our knowledge, this is the first study to examine the role of circRNA in the neuropathology of AD. We show that circRNAs impact mRNA expression by interacting with miRNA in the NAc of AD subjects. More importantly, we provide indirect evidence for the clinical importance of circRNA in the development of AUD by detecting a significant enrichment of our circRNA eQTLs among GWAS of substance abuse.
Collapse
Affiliation(s)
- Eric Vornholt
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Integrative Life Sciences Doctoral ProgramVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - John Drake
- Department of Psychiatry and Behavioral SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Mohammed Mamdani
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Gowon McMichael
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Zachary N. Taylor
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Silviu‐Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of PsychiatryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Michael F. Miles
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- VCU‐Alcohol Research CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of NeurologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Vladimir I. Vladimirov
- Virginia Institute for Psychiatric and Behavioral GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Center for Biomarker Research and Precision MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Physiology & BiophysicsVirginia Commonwealth UniversityRichmondVirginiaUSA
- School of PharmacyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Lieber Institute for Brain DevelopmentJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
18
|
Longley MJ, Lee J, Jung J, Lohoff FW. Epigenetics of alcohol use disorder-A review of recent advances in DNA methylation profiling. Addict Biol 2021; 26:e13006. [PMID: 33538087 PMCID: PMC8596445 DOI: 10.1111/adb.13006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 01/05/2021] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Alcohol use disorder (AUD) is a major contributor to morbidity and mortality worldwide. Although there is a heritable component, the etiology of AUD is complex and can involve environmental exposures like trauma and can be associated with many different patterns of alcohol consumption. Epigenetic modifications, which can mediate the influence of genetic variants and environmental variables on gene expression, have emerged as an important area of AUD research. Over the past decade, the number of studies investigating AUD and DNA methylation, a form of epigenetic modification, has grown rapidly. Yet we are still far from understanding how DNA methylation contributes to or reflects aspects of AUD. In this paper, we reviewed studies of DNA methylation and AUD and discussed how the field has evolved. We found that global DNA and candidate DNA methylation studies did not produce replicable results. To assess whether findings of epigenome-wide association studies (EWAS) were replicated, we aggregated significant findings across studies and identified 184 genes and 15 gene ontological pathways that were differentially methylated in at least two studies and four genes and three gene ontological pathways that were differentially methylated in three studies. These genes and pathways repeatedly found enrichment of immune processes, which is in line with recent developments suggesting that the immune system may be altered in AUD. Finally, we assess the current limitations of studies of DNA methylation and AUD and make recommendations on how to design future studies to resolve outstanding questions.
Collapse
Affiliation(s)
- Martha J. Longley
- Section on Clinical Genomics and Experimental TherapeuticsNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesdaMarylandUSA
| | - Jisoo Lee
- Section on Clinical Genomics and Experimental TherapeuticsNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesdaMarylandUSA
| | - Jeesun Jung
- Section on Clinical Genomics and Experimental TherapeuticsNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesdaMarylandUSA
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental TherapeuticsNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
19
|
Fedorenko OY, Mikhalitskaya EV, Toshchakova VA, Loonen AJM, Bokhan NA, Ivanova SA. Association of PIP4K2A Polymorphisms with Alcohol Use Disorder. Genes (Basel) 2021; 12:genes12101642. [PMID: 34681036 PMCID: PMC8535504 DOI: 10.3390/genes12101642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Alcohol use disorder (AUD) not only influences individuals and families but also has a lasting social impact on communities at the national level. Dopaminergic neurotransmission is involved in excessive alcohol consumption. Phosphatidylinositol-5-phosphate-4-kinase type 2 α (PIP4K2A) plays an important role in the regulation of ascending dopamine pathways. In this study; we determined possible associations between nine polymorphisms in PIP4K2A and AUD in Russian men. Methods: 279 Russian men with AUD were investigated. The control group consisted of 222 healthy men from the general Russian population. Genotyping of DNA samples for nine polymorphic variants of PIP4K2A was carried out by the Applied Biosystems™ QuantStudio™ 5 Real-Time PCR System with use of the TaqMan1 Validated SNP Genotyping Assay (Applied Biosystems; CIIIA). Results: Carriage of the PIP4K2A rs2230469*TT/T genotype/allele was a relative risk factor for developing AUD in men (p = 0.026 and p = 0.0084 accordingly). Moreover; men with AUD had a higher frequency of PIP4K2A rs746203*T allele (p = 0.023) compared to healthy men. Conclusions: For the first time; we demonstrated different PIP4K2A polymorphisms to be associated with AUD presumably due to dopamine system modulation resulting from regulation of the lateral habenula.
Collapse
Affiliation(s)
- Olga Yu. Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (E.V.M.); (V.A.T.); (N.A.B.); (S.A.I.)
- Division for Control and Diagnostics, School of Non-Destructive Testing and Security, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
- Correspondence:
| | - Ekaterina V. Mikhalitskaya
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (E.V.M.); (V.A.T.); (N.A.B.); (S.A.I.)
| | - Valentina A. Toshchakova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (E.V.M.); (V.A.T.); (N.A.B.); (S.A.I.)
| | - Anton J. M. Loonen
- PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, University of Groningen, 9713AV Groningen, The Netherlands;
| | - Nikolay A. Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (E.V.M.); (V.A.T.); (N.A.B.); (S.A.I.)
- Department of Psychotherapy and Psychological Counseling, National Research Tomsk State University, 634050 Tomsk, Russia
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (E.V.M.); (V.A.T.); (N.A.B.); (S.A.I.)
- Division for Control and Diagnostics, School of Non-Destructive Testing and Security, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| |
Collapse
|
20
|
Gender Differences among Sardinians with Alcohol Use Disorder. J Clin Med 2021; 10:jcm10204688. [PMID: 34682808 PMCID: PMC8537954 DOI: 10.3390/jcm10204688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Sardinia is an Italian island in the Mediterranean characterized by secular isolation and the singular genetic characteristics of its inhabitants. Findings obtained in populations with diverse genetic make-up and cultural background indicate gender differences and/or similarities in drinking characteristics of patients with alcohol use disorder (AUD). Knowledge of these characteristics in AUD patients is useful to improve access to treatments. This paper investigated the drinking characteristics of 66 female and 282 male outpatients with AUD, born from 1937 to 1991, living in Sardinia, and compared their characteristics with those of AUD patients living in other countries. Most Sardinian patients were men, approximately 3 years younger than women; women consumed lower amounts of alcohol than men but did not differ from men in the severity of AUD. Men were more often single than women, while a higher proportion of women reported that their mother or spouse was affected by AUD. Anxiety and depression were more prevalent among women while a higher proportion of men were affected by substance use disorders. Women were older than men at the age of first drink, regular drinking, and onset of AUD, and progressed faster than men from regular use to AUD onset. Women did not differ from men in age at first request for care, and in the lapse from AUD onset to first request for care. Women and men waited for more than 8 and 9 years, respectively, before receiving medical treatment. Gender differences progressively decreased among younger patients. Although the scarce number of women in some cohorts limits the strength of these findings, drinking characteristics of Sardinian patients did not vary significantly from those of AUD patients living in other countries. These results suggest that the number of Sardinian women with AUD is increasing and services for treatment of AUD should (a) consider women’s specific needs, and (b) realize effective policies to reduce latency prior to accessing medical treatment for both men and women with AUD.
Collapse
|
21
|
Karlsson Linnér R, Mallard TT, Barr PB, Sanchez-Roige S, Madole JW, Driver MN, Poore HE, de Vlaming R, Grotzinger AD, Tielbeek JJ, Johnson EC, Liu M, Rosenthal SB, Ideker T, Zhou H, Kember RL, Pasman JA, Verweij KJH, Liu DJ, Vrieze S, Kranzler HR, Gelernter J, Harris KM, Tucker-Drob EM, Waldman ID, Palmer AA, Harden KP, Koellinger PD, Dick DM. Multivariate analysis of 1.5 million people identifies genetic associations with traits related to self-regulation and addiction. Nat Neurosci 2021; 24:1367-1376. [PMID: 34446935 PMCID: PMC8484054 DOI: 10.1038/s41593-021-00908-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Behaviors and disorders related to self-regulation, such as substance use, antisocial behavior and attention-deficit/hyperactivity disorder, are collectively referred to as externalizing and have shared genetic liability. We applied a multivariate approach that leverages genetic correlations among externalizing traits for genome-wide association analyses. By pooling data from ~1.5 million people, our approach is statistically more powerful than single-trait analyses and identifies more than 500 genetic loci. The loci were enriched for genes expressed in the brain and related to nervous system development. A polygenic score constructed from our results predicts a range of behavioral and medical outcomes that were not part of genome-wide analyses, including traits that until now lacked well-performing polygenic scores, such as opioid use disorder, suicide, HIV infections, criminal convictions and unemployment. Our findings are consistent with the idea that persistent difficulties in self-regulation can be conceptualized as a neurodevelopmental trait with complex and far-reaching social and health correlates.
Collapse
Affiliation(s)
| | - Travis T Mallard
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Peter B Barr
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James W Madole
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Morgan N Driver
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Holly E Poore
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Ronald de Vlaming
- Department of Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Jorim J Tielbeek
- Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mengzhen Liu
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, VA CT Healthcare System, West Haven, CT, USA
| | - Rachel L Kember
- Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Mental Illness Research Education and Clinical Center, Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Joëlle A Pasman
- Behavioural Science Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Karin J H Verweij
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dajiang J Liu
- Department of Public Health Sciences, Penn State University, Hershey, PA, USA
- Institute of Personalized Medicine, Penn State University, Hershey, PA, USA
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Henry R Kranzler
- Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Mental Illness Research Education and Clinical Center, Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, VA CT Healthcare System, West Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, West Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, West Haven, CT, USA
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elliot M Tucker-Drob
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
- Center for Computational and Quantitative Genetics, Emory University, Atlanta, GA, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Philipp D Koellinger
- Department of Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA.
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
22
|
He Y, Liu H, Luo S, Amos CI, Lee JE, Yang K, Qureshi AA, Han J, Wei Q. Genetic variants of EML1 and HIST1H4E in myeloid cell-related pathway genes independently predict cutaneous melanoma-specific survival. Am J Cancer Res 2021; 11:3252-3262. [PMID: 34249459 PMCID: PMC8263692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/07/2020] [Indexed: 06/13/2023] Open
Abstract
Both in vivo and in vitro evidence has supported a key role of myeloid cells in immune suppression in melanoma and in promoting melanocytic metastases. Some single-nucleotide polymorphisms (SNPs) have been shown to predict cutaneous melanoma-specific survival (CMSS), but the association between genetic variation in myeloid cell-related genes and cutaneous melanoma (CM) patient survival remains unknown. METHODS we investigated associations between SNPs in myeloid cell-related pathway genes and CMSS in a discovery dataset of 850 CM patients and replicated the findings in another dataset of 409 CM patients. RESULTS we identified two SNPs (EML1 rs10151787 A>G and HIST1H4E rs2069018 T>C) as independent prognostic factors for CMSS, with adjusted allelic hazards ratios of 1.56 (95% confidence interval =1.19-2.05, P=0.001) and 1.66 (1.22-2.26, P=0.001), respectively; so were their combined unfavorable alleles in a dose-response manner in both discovery and replication datasets (P trend<0.001 and 0.002, respectively). Additional functional analysis revealed that both EML1 rs10151787 G and HIST1H4E rs2069018 C alleles were associated with elevated mRNA expression levels in normal tissues. CONCLUSIONS Our findings suggest that EML1 rs10151787 A>G and HIST1H4E rs2069018 T>C are independent prognostic biomarkers for CMSS.
Collapse
Affiliation(s)
- Yuanmin He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical UniversityLuzhou 646000, Sichuan, China
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of MedicineDurham, NC 27710, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer CenterHouston, TX 77030, USA
| | - Keming Yang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana UniversityIndianapolis, IN 46202, USA
| | - Abrar A Qureshi
- Department of Dermatology, Rhode Island HospitalProvidence, RI 02901, USA
- Warren Alpert Medical School at Brown UniversityProvidence, RI 02901, USA
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana UniversityIndianapolis, IN 46202, USA
- The Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBoston, MA 02115, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
- Department of Medicine, Duke University School of MedicineDurham, NC 27710, USA
| |
Collapse
|
23
|
Peng Q, Ehlers CL. Long tracks of homozygosity predict the severity of alcohol use disorders in an American Indian population. Mol Psychiatry 2021; 26:2200-2211. [PMID: 33398086 PMCID: PMC8254832 DOI: 10.1038/s41380-020-00989-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 11/20/2022]
Abstract
Runs of homozygosity (ROH) arise when an individual inherits two copies of the same haplotype segment. While ROH are ubiquitous across human populations, Native populations-with shared parental ancestry arising from isolation and endogamy-can carry a substantial enrichment for ROH. We have been investigating genetic and environmental risk factors for alcohol use disorders (AUD) in a group of American Indians (AI) who have higher rates of AUD than the general U. S. population. Here we explore whether ROH might be associated with incidence and severity of AUD in this admixed AI population (n = 742) that live on geographically contiguous reservations, using low-coverage whole genome sequences. We have found that the genomic regions in the ROH that were identified in this population had significantly elevated American Indian heritage compared with the rest of the genome. Increased ROH abundance and ROH burden are likely risk factors for AUD severity in this AI population, especially in those diagnosed with severe and moderate AUD. The association between ROH and AUD was mostly driven by ROH of moderate lengths between 1 and 2 Mb. An ROH island on chromosome 1p32.3 and a rare ROH pool on chromosome 3p12.3 were found to be significantly associated with AUD severity. They contain genes involved in lipid metabolism, oxidative stress and inflammatory responses; and OSBPL9 was found to reside on the consensus part of the ROH island. These data demonstrate that ROH are associated with risk for AUD severity in this AI population.
Collapse
Affiliation(s)
- Qian Peng
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
24
|
Rao X, Thapa KS, Chen AB, Lin H, Gao H, Reiter JL, Hargreaves KA, Ipe J, Lai D, Xuei X, Wang Y, Gu H, Kapoor M, Farris SP, Tischfield J, Foroud T, Goate AM, Skaar TC, Mayfield RD, Edenberg HJ, Liu Y. Allele-specific expression and high-throughput reporter assay reveal functional genetic variants associated with alcohol use disorders. Mol Psychiatry 2021; 26:1142-1151. [PMID: 31477794 PMCID: PMC7050407 DOI: 10.1038/s41380-019-0508-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/09/2019] [Accepted: 07/24/2019] [Indexed: 11/15/2022]
Abstract
Genome-wide association studies (GWAS) of complex traits, such as alcohol use disorders (AUD), usually identify variants in non-coding regions and cannot by themselves distinguish whether the associated variants are functional or in linkage disequilibrium with the functional variants. Transcriptome studies can identify genes whose expression differs between alcoholics and controls. To test which variants associated with AUD may cause expression differences, we integrated data from deep RNA-seq and GWAS of four postmortem brain regions from 30 subjects with AUD and 30 controls to analyze allele-specific expression (ASE). We identified 88 genes with differential ASE in subjects with AUD compared to controls. Next, to test one potential mechanism contributing to the differential ASE, we analyzed single nucleotide polymorphisms (SNPs) in the 3' untranslated regions (3'UTR) of these genes. Of the 88 genes with differential ASE, 61 genes contained 437 SNPs in the 3'UTR with at least one heterozygote among the subjects studied. Using a modified PASSPORT-seq (parallel assessment of polymorphisms in miRNA target-sites by sequencing) assay, we identified 25 SNPs that affected RNA levels in a consistent manner in two neuroblastoma cell lines, SH-SY5Y and SK-N-BE(2). Many of these SNPs are in binding sites of miRNAs and RNA-binding proteins, indicating that these SNPs are likely causal variants of AUD-associated differential ASE. In sum, we demonstrate that a combination of computational and experimental approaches provides a powerful strategy to uncover functionally relevant variants associated with the risk for AUD.
Collapse
Affiliation(s)
- Xi Rao
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kriti S Thapa
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andy B Chen
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hai Lin
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hongyu Gao
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jill L Reiter
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Katherine A Hargreaves
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joseph Ipe
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dongbing Lai
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaoling Xuei
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yue Wang
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hongmei Gu
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Manav Kapoor
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sean P Farris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Jay Tischfield
- Department of Genetics, Rutgers University, Piscataway, NJ, USA
| | - Tatiana Foroud
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Todd C Skaar
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Howard J Edenberg
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
25
|
Jensen BE, Townsley KG, Grigsby KB, Metten P, Chand M, Uzoekwe M, Tran A, Firsick E, LeBlanc K, Crabbe JC, Ozburn AR. Ethanol-Related Behaviors in Mouse Lines Selectively Bred for Drinking to Intoxication. Brain Sci 2021; 11:189. [PMID: 33557285 PMCID: PMC7915226 DOI: 10.3390/brainsci11020189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 11/18/2022] Open
Abstract
Alcohol use disorder (AUD) is a devastating psychiatric disorder that has significant wide-reaching effects on individuals and society. Selectively bred mouse lines are an effective means of exploring the genetic and neuronal mechanisms underlying AUD and such studies are translationally important for identifying treatment options. Here, we report on behavioral characterization of two replicate lines of mice that drink to intoxication, the High Drinking in the Dark (HDID)-1 and -2 mice, which have been selectively bred (20+ generations) for the primary phenotype of reaching high blood alcohol levels (BALs) during the drinking in the dark (DID) task, a binge-like drinking assay. Along with their genetically heterogenous progenitor line, Hs/Npt, we tested these mice on: DID and drinking in the light (DIL); temporal drinking patterns; ethanol sensitivity, through loss of righting reflex (LORR); and operant self-administration, including fixed ratio (FR1), fixed ratio 3:1 (FR3), extinction/reinstatement, and progressive ratio (PR). All mice consumed more ethanol during the dark than the light and both HDID lines consumed more ethanol than Hs/Npt during DIL and DID. In the dark, we found that the HDID lines achieved high blood alcohol levels early into a drinking session, suggesting that they exhibit front loading like drinking behavior in the absence of the chronicity usually required for such behavior. Surprisingly, HDID-1 (female and male) and HDID-2 (male) mice were more sensitive to the intoxicating effects of ethanol during the dark (as determined by LORR), while Hs/Npt (female and male) and HDID-2 (female) mice appeared less sensitive. We observed lower HDID-1 ethanol intake compared to either HDID-2 or Hs/Npt during operant ethanol self-administration. There were no genotype differences for either progressive ratio responding, or cue-induced ethanol reinstatement, though the latter is complicated by a lack of extinguished responding behavior. Taken together, these findings suggest that genes affecting one AUD-related behavior do not necessarily affect other AUD-related behaviors. Moreover, these findings highlight that alcohol-related behaviors can also differ between lines selectively bred for the same phenotype, and even between sexes within those same line.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Angela R. Ozburn
- Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, OR 97239, USA; (B.E.J.); (K.G.T.); (K.B.G.); (P.M.); (M.C.); (M.U.); (A.T.); (E.F.); (K.L.); (J.C.C.)
| |
Collapse
|
26
|
Li W, Thygesen JH, O'Brien NL, Heydtmann M, Smith I, Degenhardt F, Nöthen MM, Morgan MY, Bass NJ, McQuillin A. The influence of regression models on genome-wide association studies of alcohol dependence: a comparison of binary and quantitative analyses. Psychiatr Genet 2021; 31:13-20. [PMID: 33290381 DOI: 10.1097/ypg.0000000000000268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Genome-wide association studies (GWAS) of alcohol dependence syndrome (ADS) offer a platform to detect genetic risk loci. However, the majority of the ADS GWAS undertaken, to date, have utilized a case-control design and have failed to identify consistently replicable loci with the exception of protective variants within the alcohol metabolizing genes, notably ADH1B. The ADS phenotype shows considerable variability which means that the use of quantitative variables as a proxy for the severity of ADS has the potential to facilitate identification of risk loci by increasing statistical power. The current study aims to examine the influences of using binary and adjusted quantitative measures of ADS on GWAS outcomes and on calculated polygenic risk scores (PRS). METHODS A GWAS was performed in 1251 healthy controls with no history of excess alcohol use and 739 patients with ADS classified using binary DMS-IV criteria. Two additional GWAS were undertaken using a quantitative score based on DSM-IV criteria, which were applied assuming both normal and non-normal distributions of the phenotypic variables. PRS analyses were performed utilizing the data from the binary and the quantitative trait analyses. RESULTS No associations were identified at genome-wide significance in any of the individual GWAS; results were comparable in all three. The top associated single nucleotide polymorphism was located on the alcohol dehydrogenase gene cluster on chromosome 4, consistent with previous ADS GWAS. The quantitative trait analysis adjusted for the distribution of the criterion score and the associated PRS had the smallest standard errors and thus the greatest precision. CONCLUSION Further exploitation of the use of qualitative trait analysis in GWAS in ADS is warranted.
Collapse
Affiliation(s)
- Wenqianglong Li
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London
| | - Johan Hilge Thygesen
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London
| | - Niamh Louise O'Brien
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London
| | - Mathis Heydtmann
- Royal Alexandria Hospital, NHS Greater Glasgow and Clyde, Paisley
| | - Iain Smith
- Glasgow Addiction Services, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn
- Department of Child and Adolescent Psychiatry, University of Duisburg-Essen, Essen, Germany
| | - Markus Maria Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn
| | - Marsha Yvonne Morgan
- UCL Institute for Liver & Digestive Health, Division of Medicine, Royal Free Campus, University College London, London, UK
| | - Nicholas James Bass
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London
| | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London
| |
Collapse
|
27
|
Corlin L, Liu C, Lin H, Leone D, Yang Q, Ngo D, Levy D, Cupples LA, Gerszten RE, Larson MG, Vasan RS. Proteomic Signatures of Lifestyle Risk Factors for Cardiovascular Disease: A Cross-Sectional Analysis of the Plasma Proteome in the Framingham Heart Study. J Am Heart Assoc 2021; 10:e018020. [PMID: 33372532 PMCID: PMC7955453 DOI: 10.1161/jaha.120.018020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Background Proteomic biomarkers related to cardiovascular disease risk factors may offer insights into the pathogenesis of cardiovascular disease. We investigated whether modifiable lifestyle risk factors for cardiovascular disease are associated with distinctive proteomic signatures. Methods and Results We analyzed 1305 circulating plasma proteomic biomarkers (assayed using the SomaLogic platform) in 897 FHS (Framingham Heart Study) Generation 3 participants (mean age 46±8 years; 56% women; discovery sample) and 1121 FOS (Framingham Offspring Study) participants (mean age 52 years; 54% women; validation sample). Participants were free of hypertension, diabetes mellitus, and clinical cardiovascular disease. We used linear mixed effects models (adjusting for age, sex, body mass index, and family structure) to relate levels of each inverse-log transformed protein to 3 lifestyle factors (ie, smoking, alcohol consumption, and physical activity). A Bonferroni-adjusted P value indicated statistical significance (based on number of proteins and traits tested, P<4.2×10-6 in the discovery sample; P<6.85×10-4 in the validation sample). We observed statistically significant associations of 60 proteins with smoking (37/40 top proteins validated in FOS), 30 proteins with alcohol consumption (23/30 proteins validated), and 5 proteins with physical activity (2/3 proteins associated with the physical activity index validated). We assessed the associations of protein concentrations with previously identified genetic variants (protein quantitative trait loci) linked to lifestyle-related disease traits in the genome-wide-association study catalogue. The protein quantitative trait loci were associated with coronary artery disease, inflammation, and age-related mortality. Conclusions Our cross-sectional study from a community-based sample elucidated distinctive sets of proteins associated with 3 key lifestyle factors.
Collapse
Affiliation(s)
- Laura Corlin
- Boston University Department of MedicineBostonMA
- Department of Public Health and Community MedicineTufts University School of MedicineBostonMA
- Department of Civil and Environmental EngineeringTufts University School of EngineeringMedfordMA
| | - Chunyu Liu
- Boston University School of Public HealthBostonMA
| | | | | | - Qiong Yang
- Boston University School of Public HealthBostonMA
| | - Debby Ngo
- Department of MedicineBeth Israel Deaconess Medical CenterBostonMA
- Harvard Medical SchoolBostonMA
| | - Daniel Levy
- Population Sciences BranchNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMD
- Framingham Heart StudyFraminghamMA
| | - L. Adrienne Cupples
- Boston University School of Public HealthBostonMA
- Framingham Heart StudyFraminghamMA
| | - Robert E. Gerszten
- Department of MedicineBeth Israel Deaconess Medical CenterBostonMA
- Harvard Medical SchoolBostonMA
| | - Martin G. Larson
- Boston University School of Public HealthBostonMA
- Framingham Heart StudyFraminghamMA
| | - Ramachandran S. Vasan
- Boston University Department of MedicineBostonMA
- Boston University School of Public HealthBostonMA
- Framingham Heart StudyFraminghamMA
- Boston University Center for Computing and Data SciencesBostonMA
| |
Collapse
|
28
|
Wang K, Duan Y, Duan W, Yu Y, Zheng N, Hu J, He J, Chen H, Liang M. Bibliometric Insights in Genetic Factors of Substance-Related Disorders: Intellectual Developments, Turning Points, and Emerging Trends. Front Psychiatry 2021; 12:620489. [PMID: 34135780 PMCID: PMC8200466 DOI: 10.3389/fpsyt.2021.620489] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Substance-related disorders are a group of medical conditions that affect a person's brain and behavior and lead to an inability to control the use of legal or illegal drug(s) or medication. Substance-related disorder is a serious public health and society problem worldwide. Genetic factors have been proven to have an important role. Researchers have carried out a lot of work in this field, and a large number of research results have been published in academic journals around the world. However, there are few overviews of research progress, presentation, and development trends in this field. In this study, a total of 636 articles related to genetic factors of substance-related disorders were retrieved from the Web of Science (WoS) database from 1997 to 2018, and the scientific literatures were analyzed by bibliometrics. The study found that the United States (US) has maintained a leading position in the field of research, with many core institutions and plenty of high-quality research results. Alcohol use disorder is still the most concerning issue in this field. Over the past 20 years, new techniques such as genome-wide association study (GWAS) based on high-throughput sequencing technology have replaced family studies, twin studies, and retrospective studies in this field. We believe that it is urgent to study the genetic factors of substance-related disorders, which can greatly deepen the understanding of the pathogenesis of substance-related disorders and may provide potential targets for precise treatment of such diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijie Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicheng Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxin Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Zheng
- Department of Pathology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jin Hu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia He
- Department of Public Health, Shihezi University School of Medicine, Shihezi, China
| | - Haihong Chen
- School of Health Policy and Management, Nanjing Medical University, Nanjing, China
| | - Man Liang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Thapa KS, Chen AB, Lai D, Xuei X, Wetherill L, Tischfield JA, Liu Y, Edenberg HJ. Identification of Functional Genetic Variants Associated With Alcohol Dependence and Related Phenotypes Using a High-Throughput Assay. Alcohol Clin Exp Res 2020; 44:2494-2518. [PMID: 33119910 PMCID: PMC7725989 DOI: 10.1111/acer.14492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/20/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Genome-wide association studies (GWAS) of alcohol dependence (AD) and related phenotypes have identified multiple loci, but the functional variants underlying the loci have in most cases not been identified. Noncoding variants can influence phenotype by affecting gene expression; for example, variants in the 3' untranslated regions (3'UTR) can affect gene expression posttranscriptionally. METHODS We adapted a high-throughput assay known as PASSPORT-seq (parallel assessment of polymorphisms in miRNA target sites by sequencing) to identify among variants associated with AD and related phenotypes those that cause differential expression in neuronal cell lines. Based upon meta-analyses of alcohol-related traits in African American and European Americans in the Collaborative Study on the Genetics of Alcoholism, we tested 296 single nucleotide polymorphisms (SNPs with meta-analysis p values ≤ 0.001) that were located in 3'UTRs. RESULTS We identified 60 SNPs that affected gene expression (false discovery rate [FDR] < 0.05) in SH-SY5Y cells and 92 that affected expression in SK-N-BE(2) cells. Among these, 30 SNPs altered RNA levels in the same direction in both cell lines. Many of these SNPs reside in the binding sites of miRNAs and RNA-binding proteins and are expression quantitative trait loci of genes including KIF6,FRMD4A,CADM2,ADD2,PLK2, and GAS7. CONCLUSION The SNPs identified in the PASSPORT-seq assay are functional variants that might affect the risk for AD and related phenotypes. Our study provides insights into gene regulation in AD and demonstrates the value of PASSPORT-seq as a tool to screen genetic variants in GWAS loci for one potential mechanism of action.
Collapse
Affiliation(s)
- Kriti S. Thapa
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andy B Chen
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Dongbing Lai
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiaoling Xuei
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leah Wetherill
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jay A. Tischfield
- Department of Genetics, Rutgers University, Piscataway, NJ, 99999, USA
| | - Yunlong Liu
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Howard J. Edenberg
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
30
|
Parker CC, Lusk R, Saba LM. Alcohol Sensitivity as an Endophenotype of Alcohol Use Disorder: Exploring Its Translational Utility between Rodents and Humans. Brain Sci 2020; 10:E725. [PMID: 33066036 PMCID: PMC7600833 DOI: 10.3390/brainsci10100725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022] Open
Abstract
Alcohol use disorder (AUD) is a complex, chronic, relapsing disorder with multiple interacting genetic and environmental influences. Numerous studies have verified the influence of genetics on AUD, yet the underlying biological pathways remain unknown. One strategy to interrogate complex diseases is the use of endophenotypes, which deconstruct current diagnostic categories into component traits that may be more amenable to genetic research. In this review, we explore how an endophenotype such as sensitivity to alcohol can be used in conjunction with rodent models to provide mechanistic insights into AUD. We evaluate three alcohol sensitivity endophenotypes (stimulation, intoxication, and aversion) for their translatability across human and rodent research by examining the underlying neurobiology and its relationship to consumption and AUD. We show examples in which results gleaned from rodents are successfully integrated with information from human studies to gain insight in the genetic underpinnings of AUD and AUD-related endophenotypes. Finally, we identify areas for future translational research that could greatly expand our knowledge of the biological and molecular aspects of the transition to AUD with the broad hope of finding better ways to treat this devastating disorder.
Collapse
Affiliation(s)
- Clarissa C. Parker
- Department of Psychology and Program in Neuroscience, Middlebury College, Middlebury, VT 05753, USA
| | - Ryan Lusk
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Laura M. Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
31
|
Lathen DR, Merrill CB, Rothenfluh A. Flying Together: Drosophila as a Tool to Understand the Genetics of Human Alcoholism. Int J Mol Sci 2020; 21:E6649. [PMID: 32932795 PMCID: PMC7555299 DOI: 10.3390/ijms21186649] [Citation(s) in RCA: 10] [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: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Alcohol use disorder (AUD) exacts an immense toll on individuals, families, and society. Genetic factors determine up to 60% of an individual's risk of developing problematic alcohol habits. Effective AUD prevention and treatment requires knowledge of the genes that predispose people to alcoholism, play a role in alcohol responses, and/or contribute to the development of addiction. As a highly tractable and translatable genetic and behavioral model organism, Drosophila melanogaster has proven valuable to uncover important genes and mechanistic pathways that have obvious orthologs in humans and that help explain the complexities of addiction. Vinegar flies exhibit remarkably strong face and mechanistic validity as a model for AUDs, permitting many advancements in the quest to understand human genetic involvement in this disease. These advancements occur via approaches that essentially fall into one of two categories: (1) discovering candidate genes via human genome-wide association studies (GWAS), transcriptomics on post-mortem tissue from AUD patients, or relevant physiological connections, then using reverse genetics in flies to validate candidate genes' roles and investigate their molecular function in the context of alcohol. (2) Utilizing flies to discover candidate genes through unbiased screens, GWAS, quantitative trait locus analyses, transcriptomics, or single-gene studies, then validating their translational role in human genetic surveys. In this review, we highlight the utility of Drosophila as a model for alcoholism by surveying recent advances in our understanding of human AUDs that resulted from these various approaches. We summarize the genes that are conserved in alcohol-related function between humans and flies. We also provide insight into some advantages and limitations of these approaches. Overall, this review demonstrates how Drosophila have and can be used to answer important genetic questions about alcohol addiction.
Collapse
Affiliation(s)
- Daniel R. Lathen
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
| | - Collin B. Merrill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
32
|
Scarnati MS, Boreland AJ, Joel M, Hart RP, Pang ZP. Differential sensitivity of human neurons carrying μ opioid receptor (MOR) N40D variants in response to ethanol. Alcohol 2020; 87:97-109. [PMID: 32561311 PMCID: PMC7958146 DOI: 10.1016/j.alcohol.2020.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022]
Abstract
The acute and chronic effects of alcohol on the brain and behavior are linked to alterations in inhibitory synaptic transmission. Alcohol's most consistent effect at the synaptic level is probably a facilitation of γ-aminobutyric acid (GABA) release, as seen from several rodent studies. The impact of alcohol on GABAergic neurotransmission in human neurons is unknown, due to a lack of a suitable experimental model. Human neurons can also be used to model effects of genetic variants linked with alcohol use disorders (AUDs). The A118G single nucleotide polymorphism (SNP rs1799971) of the OPRM1 gene encoding the N40D (D40 minor allele) mu-opioid receptor (MOR) variant has been linked with individuals who have an AUD. However, while N40D is clearly associated with other drugs of abuse, involvement with AUDs is controversial. In this study, we employed Ascl1-and Dlx2-induced inhibitory neuronal cells (AD-iNs) generated from human iPS cell lines carrying N40D variants, and investigated the impact of ethanol acutely and chronically on GABAergic synaptic transmission. We found that N40 AD-iNs display a stronger facilitation (versus D40) of spontaneous and miniature inhibitory postsynaptic current frequency in response to acute ethanol application. Quantitative immunocytochemistry of Synapsin 1+ synaptic puncta revealed a similar synapse number between N40 and D40 iNs, suggesting an ethanol modulation of presynaptic GABA release without affecting synapse density. Interestingly, D40 iNs exposed to chronic intermittent ethanol application caused a significant increase in mIPSC frequency, with only a modest enhancement observed in N40 iNs. These data suggest that the MOR genotype may confer differential sensitivity to synaptic output, which depends on ethanol exposure time and concentration for AD-iNs and may help explain alcohol dependence in individuals who carry the MOR D40 SNPs. Furthermore, this study supports the use of human neuronal cells carrying risk-associated genetic variants linked to disease, as in vitro models to assay the synaptic actions of alcohol on human neuronal cells.
Collapse
Affiliation(s)
- Matthew S Scarnati
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Andrew J Boreland
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Marisa Joel
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA; Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA; Human Genetics Institute of New Jersey, Piscataway, NJ, 08854, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
33
|
The Family Check-up Intervention Moderates Polygenic Influences on Long-Term Alcohol Outcomes: Results from a Randomized Intervention Trial. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2020; 20:975-985. [PMID: 31175564 DOI: 10.1007/s11121-019-01024-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alcohol problems are influenced by both genetic and environmental factors. Evidence from twin models and measured gene-environment interaction studies has demonstrated that the importance of genetic influences changes as a function of the environment. Research has also shown that family-centered interventions may protect genetically susceptible youth from developing substance use problems. In this study, we brought large-scale gene identification findings into an intervention study to examine gene-by-intervention effects. Using genome-wide polygenic scores derived from an independent genome-wide association study of adult alcohol dependence, we examined whether an adolescent family-centered intervention would moderate the effect of genetic risk for alcohol dependence on lifetime alcohol dependence in young adulthood, approximately 15 years after the start of intervention, among European American (N = 271; 48.3% in the intervention condition) and African American individuals (N = 192; 51.6% in the intervention condition). We found that among European American individuals, the intervention moderated the association between alcohol dependence polygenic scores and lifetime alcohol dependence diagnosis in young adulthood. Among participants in the control condition, higher alcohol dependence polygenic scores were associated with a greater likelihood of receiving an alcohol dependence diagnosis; in contrast, among participants in the intervention condition, there was no association between alcohol dependence polygenic scores and alcohol dependence diagnosis. No moderation effect was found among African Americans. These results demonstrate that modifying environments of genetically vulnerable youth could reduce the likelihood of developing alcohol dependence and underscore the significance of environmentally focused prevention and intervention efforts.
Collapse
|
34
|
Bellia F, Fernández MS, Fabio MC, Pucci M, Pautassi RM, D'Addario C. Selective alterations in endogenous opioid system genes expression in rats selected for high ethanol intake during adolescence. Drug Alcohol Depend 2020; 212:108025. [PMID: 32442753 DOI: 10.1016/j.drugalcdep.2020.108025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/31/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
Historically, the roots of alcoholism have been linked to either environment or heredity. However, the interaction between these factors is still largely unexplored. The evidence supports a link between alcohol consumption and the endogenous opioid system. We here studied the opioid genes expression in male and female Wistar rats derived from a short-term breeding program which selected -- at adolescence -- for high (ADHI line) or low (ADLO line) ethanol drinking. Specifically, in this work we analyzed central opioid gene expression in the rats of the second filial generation (S2-ADLO and S2-ADHI). Selective downregulation of pronociceptin (Pnoc) and its receptor (Oprl1) mRNA levels were observed in the prefrontal cortex of male S2-ADHI rats when compared to S2-ADLO, and for Oprl1 also in the nucleus accumbens. An increase in gene expression was instead observed for pro-opiomelanocortin (Pomc) in the nucleus accumbens of S2-ADHI males when compared to S2-ADLO, as well as for mu opioid receptor (Oprm1) but in females. The differences in mRNA levels may be due to the different alcohol consumption between the two groups of rats or may represent pre-existing differences between them. Moreover, we show a sex-specific modulation of the expression of these genes, thus pointing out the importance of sex on ethanol responses. The results might lead to more specific and effective pharmacological treatments for alcoholism.
Collapse
Affiliation(s)
| | - Macarena Soledad Fernández
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Carolina Fabio
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - Claudio D'Addario
- Università degli Studi di Teramo, Teramo, Italy; Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden.
| |
Collapse
|
35
|
Bauer LO, Covault JM. GRM8 genotype is associated with externalizing disorders and greater inter-trial variability in brain activation during a response inhibition task. Clin Neurophysiol 2020; 131:1180-1186. [PMID: 32299001 PMCID: PMC7198333 DOI: 10.1016/j.clinph.2020.02.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/13/2020] [Accepted: 02/25/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The present investigation tested the association of a novel measure of brain activation recorded during a simple motor inhibition task with a GRM8 genetic locus implicated in risk for substance dependence. METHODS 122 European-American adults were genotyped at rs1361995 and evaluated against DSM-IV criteria for Alcohol Dependence, Cocaine Dependence, Conduct Disorder, and Antisocial Personality Disorder. Also, their brain activity was recorded in response to rare, so-called "No-Go" stimuli presented during a continuous performance test. Brain activity was quantified with two indices: (1) the amplitude of the No-Go P300 electroencephalographic response averaged across trials; and (2) the inter-trial variability of the response. RESULTS The absence of the minor allele at the candidate locus was associated with all of the evaluated diagnoses. In comparison to minor allele carriers, major allele homozygotes also demonstrated increased inter-trial variability in No-Go P300 response amplitude but no difference in average amplitude. CONCLUSIONS GRM8 genotype is associated with Alcohol and Cocaine Dependence as well as personality risk factors for dependence. The association may be mediated through an inherited instability in brain function that affects cognitive control. SIGNIFICANCE The present study focuses on a metric and brain mechanism not typically considered or theorized in studies of patients with substance use disorders.
Collapse
Affiliation(s)
- Lance O Bauer
- Department of Psychiatry, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-1410, USA.
| | - Jonathan M Covault
- Department of Psychiatry, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-1410, USA
| |
Collapse
|
36
|
Lim AC, Green R, Grodin EN, Venegas A, Meredith LR, Donato S, Burnette E, Ray LA. Alcohol Cue-Induced Ventral Striatum Activity Predicts Subsequent Alcohol Self-Administration. Alcohol Clin Exp Res 2020; 44:1224-1233. [PMID: 32406553 PMCID: PMC7336863 DOI: 10.1111/acer.14342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Human laboratory paradigms are a pillar in medication development for alcohol use disorders (AUD). Neuroimaging paradigms, in which individuals are exposed to cues that elicit neural correlates of alcohol craving (e.g., mesocorticolimbic activation), are increasingly utilized to test the effects of AUD medications. Elucidation of the translational effects of these neuroimaging paradigms on human laboratory paradigms, such as self-administration, is warranted. The current study is a secondary analysis examining whether alcohol cue-induced activation in the ventral striatum is predictive of subsequent alcohol self-administration in the laboratory. METHODS Non-treatment-seeking heavy drinkers of East Asian descent (n = 41) completed a randomized, placebo-controlled, double-blind, crossover experiment on the effects of naltrexone on neuroimaging and human laboratory paradigms. Participants completed 5 days of study medication (or placebo); on day 4, they completed a neuroimaging alcohol taste cue-reactivity task. On the following day (day 5), participants completed a 60-minute alcohol self-administration paradigm. RESULTS Multilevel Cox regressions indicated a significant effect of taste cue-elicited ventral striatum activation on latency to first drink, Wald χ2 = 2.88, p = 0.05, such that those with higher ventral striatum activation exhibited shorter latencies to consume their first drink. Similarly, ventral striatum activation was positively associated with total number of drinks consumed, F(1, 38) = 5.90, p = 0.02. These effects were significant after controlling for alcohol use severity, OPRM1 genotype, and medication. Other potential regions of interest (anterior cingulate, thalamus) were not predictive of self-administration outcomes. CONCLUSIONS Neuroimaging alcohol taste cue paradigms may be predictive of laboratory paradigms such as self-administration. Elucidation of the relationships among different paradigms will inform how these paradigms may be used synergistically in experimental medicine and medication development.
Collapse
Affiliation(s)
- Aaron C. Lim
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - ReJoyce Green
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erica N. Grodin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexandra Venegas
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lindsay R. Meredith
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Suzanna Donato
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elizabeth Burnette
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lara A. Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
37
|
Sanchez-Roige S, Palmer AA, Clarke TK. Recent Efforts to Dissect the Genetic Basis of Alcohol Use and Abuse. Biol Psychiatry 2020; 87:609-618. [PMID: 31733789 PMCID: PMC7071963 DOI: 10.1016/j.biopsych.2019.09.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/14/2019] [Accepted: 09/13/2019] [Indexed: 01/29/2023]
Abstract
Alcohol use disorder (AUD) is defined by several symptom criteria, which can be dissected further at the genetic level. Over the past several years, our understanding of the genetic factors influencing alcohol use and abuse has progressed tremendously; numerous loci have been implicated in different aspects of alcohol use. Previously known associations with alcohol-metabolizing enzymes (ADH1B, ALDH2) have been replicated definitively. In addition, novel associations with loci containing the genes KLB, GCKR, CRHR1, and CADM2 have been reported. Downstream analyses have leveraged these genetic findings to reveal important relationships between alcohol use behaviors and both physical and mental health. AUD and aspects of alcohol misuse have been shown to overlap strongly with psychiatric disorders, whereas aspects of alcohol consumption have shown stronger links to metabolism. These results demonstrate that the genetic architecture of alcohol consumption only partially overlaps with the genetics of clinically defined AUD. We discuss the limitations of using quantitative measures of alcohol use as proxy measures for AUD, and we outline how future studies will require careful phenotype harmonization to properly capture the genetic liability to AUD.
Collapse
Affiliation(s)
- Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, California.
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, California; Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Toni-Kim Clarke
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Ning K, Zhao L, Matloff W, Sun F, Toga AW. Association of relative brain age with tobacco smoking, alcohol consumption, and genetic variants. Sci Rep 2020; 10:10. [PMID: 32001736 PMCID: PMC6992742 DOI: 10.1038/s41598-019-56089-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/06/2019] [Indexed: 12/24/2022] Open
Abstract
Brain age is a metric that quantifies the degree of aging of a brain based on whole-brain anatomical characteristics. While associations between individual human brain regions and environmental or genetic factors have been investigated, how brain age is associated with those factors remains unclear. We investigated these associations using UK Biobank data. We first trained a statistical model for obtaining relative brain age (RBA), a metric describing a subject's brain age relative to peers, based on whole-brain anatomical measurements, from training set subjects (n = 5,193). We then applied this model to evaluation set subjects (n = 12,115) and tested the association of RBA with tobacco smoking, alcohol consumption, and genetic variants. We found that daily or almost daily consumption of tobacco and alcohol were both significantly associated with increased RBA (P < 0.001). We also found SNPs significantly associated with RBA (p-value < 5E-8). The SNP most significantly associated with RBA is located in MAPT gene. Our results suggest that both environmental and genetic factors are associated with structural brain aging.
Collapse
Affiliation(s)
- Kaida Ning
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, 90033, USA
- Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Lu Zhao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, 90033, USA
| | - Will Matloff
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Fengzhu Sun
- Molecular and Computational Biology Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Arthur W Toga
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, 90033, USA.
| |
Collapse
|
39
|
Nieto SJ, Grodin EN, Ray LA. On the path towards personalized medicine: Implications of pharmacogenetic studies of alcohol use disorder medications. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:43-54. [PMID: 34291172 DOI: 10.1080/23808993.2020.1724510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction The heritability of alcohol use disorder (AUD) is estimated to be ~50%; however, the genetic basis of the disease is still poorly understood. The genetic variants identified thus far only explain a small percentage of AUD phenotypic variability. While genome-wide association studies (GWAS) are impacted by technical and methodological limitations, genetic variants that have been identified independently of GWAS findings can moderate the efficacy of AUD medications. Areas Covered This review discusses findings from clinical pharmacogenetic studies of AUD medications. While the pharmacogenetic studies reviewed involve several genetic variants in the major neurotransmitter systems, genetic loci in the opioid system have garnered the most attention. Expert Opinion The clinical utility of pharmacogenetics in AUD populations is uncertain at this time. There are several ongoing prospective clinical trials that will enhance knowledge regarding the applicability of pharmacogenetics in clinical populations. We recommend that future work in this area consider reverse translating from genotype to phenotype, mapping genes to stages of the addiction cycle, mapping genes to neural circuits, and harnessing large population-based cohorts.
Collapse
Affiliation(s)
- Steven J Nieto
- University of California Los Angeles, Department of Psychology, Los Angeles, CA, USA
| | - Erica N Grodin
- University of California Los Angeles, Department of Psychology, Los Angeles, CA, USA
| | - Lara A Ray
- University of California Los Angeles, Department of Psychology, Los Angeles, CA, USA.,University of California, Los Angeles, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA.,University of California Los Angeles, Brain Research Institute, Los Angeles, CA, USA
| |
Collapse
|
40
|
Ksinan AJ, Su J, Aliev F, Dick DM. Unpacking Genetic Risk Pathways for College Student Alcohol Consumption: The Mediating Role of Impulsivity. Alcohol Clin Exp Res 2019; 43:2100-2110. [PMID: 31373688 PMCID: PMC6779491 DOI: 10.1111/acer.14157] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/18/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND The period of college represents a particularly risky developmental stage with regard to alcohol use, as college students engage in more risky drinking behaviors than their noncollege peers, and such problematic alcohol use is associated with far-reaching negative consequences. Existing findings from genome-wide association studies (GWAS) indicate that alcohol consumption has a complex polygenic etiology. Currently, there is a lack of studies examining genetic risk for alcohol consumption using polygenic risk scores (PRS) in college samples. In this study, we examined whether alcohol-specific and risky behavior-related PRS were longitudinally associated with alcohol consumption among college students and whether this effect might be partially mediated by impulsivity domains. METHODS The sample included n = 2,385 European ancestry (EA) and n = 1,153 African ancestry (AA) college students assessed over the course of 4 years. To indicate genetic risk, 2 PRS were created based on recent large-scale GWAS: alcohol consumption (Liu et al., 2019) -drinks per week (DPW)-PRS and risky behaviors (Linnér et al., 2019) -RISK-PRS. The main outcome was alcohol consumption, measured across 4 waves of follow-up data. The UPPS-P impulsivity subscales were examined as mediators of the genetic effect on alcohol consumption. RESULTS The results from structural equation modeling showed that among EA students, both DPW-PRS and RISK-PRS had significant positive effects on alcohol consumption above and beyond UPPS dimensions and control variables. RISK-PRS explained larger portion of variance in alcohol consumption than DPW-PRS. RISK-PRS showed a significant indirect effect on alcohol consumption through sensation seeking and lack of perseverance; no significant indirect effect of DPW-PRS was found. No significant association of either PRS or alcohol consumption was found for AA participants. CONCLUSIONS The current results found that PRS related to more broadly defined risky behaviors predicted alcohol consumption across college years and that this association was partially mediated via dimensions of impulsivity.
Collapse
Affiliation(s)
- Albert J. Ksinan
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA 23284
| | - Jinni Su
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA 23284
| | - Fazil Aliev
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA 23284
| | - Spit for Science Workgroup
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA 23284
- The Spit for Science Working Group: Spit for Science
Director: Danielle M. Dick. Registry management: Kimberly Pedersen, Zoe Neale,
Nathaniel Thomas. Data cleaning and management: Amy E. Adkins, Nathaniel Thomas, Zoe
Neale, Kimberly Pedersen, Thomas Bannard & Seung B. Cho. Data collection: Amy E.
Adkins, Peter Barr, Erin C. Berenz, Erin Caraway, Seung B. Cho, James S. Clifford,
Megan Cooke, Elizabeth Do, Alexis C. Edwards, Neeru Goyal, Laura M. Hack, Lisa J.
Halberstadt, Sage Hawn, Sally Kuo, Emily Lasko, Jennifer Lend, Mackenzie Lind,
Elizabeth Long, Alexandra Martelli, Jacquelyn L. Meyers, Kerry Mitchell, Ashlee
Moore, Arden Moscati, Aashir Nasim, Zoe Neale, Jill Opalesky, Cassie Overstreet, A.
Christian Pais, Kimberly Pedersen, Tarah Raldiris, Jessica Salvatore, Jeanne Savage,
Rebecca Smith, David Sosnowski, Jinni Su, Nathaniel Thomas, Chloe Walker, Marcie
Walsh, Teresa Willoughby, Madison Woodroof & Jia Yan. Genotypic data processing
and cleaning: Cuie Sun, Brandon Wormley, Brien Riley, Fazil Aliev, Roseann Peterson
& Bradley T. Webb
| | - Danielle M. Dick
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA 23284
| |
Collapse
|
41
|
Bogenpohl JW, Smith ML, Farris SP, Dumur CI, Lopez MF, Becker HC, Grant KA, Miles MF. Cross-Species Co-analysis of Prefrontal Cortex Chronic Ethanol Transcriptome Responses in Mice and Monkeys. Front Mol Neurosci 2019; 12:197. [PMID: 31456662 PMCID: PMC6701453 DOI: 10.3389/fnmol.2019.00197] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Despite recent extensive genomic and genetic studies on behavioral responses to ethanol, relatively few new therapeutic targets for the treatment of alcohol use disorder have been validated. Here, we describe a cross-species genomic approach focused on identifying gene networks associated with chronic ethanol consumption. To identify brain mechanisms underlying a chronic ethanol consumption phenotype highly relevant to human alcohol use disorder, and to elucidate potential future therapeutic targets, we conducted a genomic study in a non-human primate model of chronic open-access ethanol consumption. Microarray analysis of RNA expression in anterior cingulate and subgenual cortices from rhesus macaques was performed across multiple cohorts of animals. Gene networks correlating with ethanol consumption or showing enrichment for ethanol-regulated genes were identified, as were major ethanol-related hub genes within these networks. A subsequent consensus module analysis was used to co-analyze monkey data with expression data from a chronic intermittent ethanol vapor-exposure and consumption model in C57BL/6J mice. Ethanol-related gene networks conserved between primates and rodents were enriched for genes involved in discrete biological functions, including; myelination, synaptic transmission, chromatin modification, Golgi apparatus function, translation, cellular respiration, and RNA processing. The myelin-related network, in particular, showed strong correlations with ethanol consumption behavior and displayed marked network reorganization between control and ethanol-drinking animals. Further bioinformatics analysis revealed that these networks also showed highly significant overlap with other ethanol-regulated gene sets. Altogether, these studies provide robust primate and rodent cross-species validation of gene networks associated with chronic ethanol consumption. Our results also suggest potential novel focal points for future therapeutic interventions in alcohol use disorder.
Collapse
Affiliation(s)
- James W Bogenpohl
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, United States
| | - Maren L Smith
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Sean P Farris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, United States
| | - Catherine I Dumur
- Aurora Diagnostics-Sonic Healthcare, Bernhardt Laboratories, Jacksonville, FL, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kathleen A Grant
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States.,Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States.,VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
42
|
Lai D, Wetherill L, Bertelsen S, Carey CE, Kamarajan C, Kapoor M, Meyers JL, Anokhin AP, Bennett DA, Bucholz KK, Chang KK, De Jager PL, Dick DM, Hesselbrock V, Kramer J, Kuperman S, Nurnberger JI, Raj T, Schuckit M, Scott DM, Taylor RE, Tischfield J, Hariri AR, Edenberg HJ, Agrawal A, Bogdan R, Porjesz B, Goate AM, Foroud T. Genome-wide association studies of alcohol dependence, DSM-IV criterion count and individual criteria. GENES, BRAIN, AND BEHAVIOR 2019; 18:e12579. [PMID: 31090166 PMCID: PMC6612573 DOI: 10.1111/gbb.12579] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/19/2019] [Accepted: 05/11/2019] [Indexed: 01/04/2023]
Abstract
Genome-wide association studies (GWAS) of alcohol dependence (AD) have reliably identified variation within alcohol metabolizing genes (eg, ADH1B) but have inconsistently located other signals, which may be partially attributable to symptom heterogeneity underlying the disorder. We conducted GWAS of DSM-IV AD (primary analysis), DSM-IV AD criterion count (secondary analysis), and individual dependence criteria (tertiary analysis) among 7418 (1121 families) European American (EA) individuals from the Collaborative Study on the Genetics of Alcoholism (COGA). Trans-ancestral meta-analyses combined these results with data from 3175 (585 families) African-American (AA) individuals from COGA. In the EA GWAS, three loci were genome-wide significant: rs1229984 in ADH1B for AD criterion count (P = 4.16E-11) and Desire to cut drinking (P = 1.21E-11); rs188227250 (chromosome 8, Drinking more than intended, P = 6.72E-09); rs1912461 (chromosome 15, Time spent drinking, P = 1.77E-08). In the trans-ancestral meta-analysis, rs1229984 was associated with multiple phenotypes and two additional loci were genome-wide significant: rs61826952 (chromosome 1, DSM-IV AD, P = 8.42E-11); rs7597960 (chromosome 2, Time spent drinking, P = 1.22E-08). Associations with rs1229984 and rs18822750 were replicated in independent datasets. Polygenic risk scores derived from the EA GWAS of AD predicted AD in two EA datasets (P < .01; 0.61%-1.82% of variance). Identified novel variants (ie, rs1912461, rs61826952) were associated with differential central evoked theta power (loss - gain; P = .0037) and reward-related ventral striatum reactivity (P = .008), respectively. This study suggests that studying individual criteria may unveil new insights into the genetic etiology of AD liability.
Collapse
Affiliation(s)
- Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN
| | - Sarah Bertelsen
- Department of Neuroscience, Icahn School of Medicine at Mt.
Sinai, New York, NY
| | - Caitlin E. Carey
- BRAIN Lab, Department of Psychological and Brain Sciences,
Washington University School of Medicine, St. Louis, MO
| | - Chella Kamarajan
- Henri Begleiter Neurodynamics Lab, Department of
Psychiatry, State University of New York, Downstate Medical Center, Brooklyn,
NY
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mt.
Sinai, New York, NY
| | - Jacquelyn L. Meyers
- Henri Begleiter Neurodynamics Lab, Department of
Psychiatry, State University of New York, Downstate Medical Center, Brooklyn,
NY
| | - Andrey P. Anokhin
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, MO
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, IL
| | - Kathleen K. Bucholz
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, MO
| | - Katharine K. Chang
- BRAIN Lab, Department of Psychological and Brain Sciences,
Washington University School of Medicine, St. Louis, MO
| | - Philip L. De Jager
- Departments of Neurology and Psychiatry, Brigham and
Women's Hospital, Boston, MA
| | - Danielle M. Dick
- Department of Psychology, Virginia Commonwealth University,
Richmond, VA
| | | | - John Kramer
- Department of Psychiatry, Roy Carver College of Medicine,
University of Iowa, Iowa City, IA
| | - Samuel Kuperman
- Department of Psychiatry, Roy Carver College of Medicine,
University of Iowa, Iowa City, IA
| | - John I. Nurnberger
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN
- Department of Psychiatry, Indiana University School of
Medicine, Indianapolis, IN
| | - Towfique Raj
- Department of Neuroscience, Icahn School of Medicine at Mt.
Sinai, New York, NY
| | - Marc Schuckit
- Department of Psychiatry, University of California, San
Diego Medical School, San Diego, CA
| | - Denise M. Scott
- Departments of Pediatrics and Human Genetics, Howard
University, Washington, DC
| | | | | | - Ahmad R. Hariri
- Laboratory of NeuroGenetics, Department of Psychology and
Neuroscience, Duke University, Durham, NC, USA
| | - Howard J. Edenberg
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana
University School of Medicine, Indianapolis, IN
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, MO
| | - Ryan Bogdan
- BRAIN Lab, Department of Psychological and Brain Sciences,
Washington University School of Medicine, St. Louis, MO
| | - Bernice Porjesz
- Henri Begleiter Neurodynamics Lab, Department of
Psychiatry, State University of New York, Downstate Medical Center, Brooklyn,
NY
| | - Alison M. Goate
- Department of Neuroscience, Icahn School of Medicine at Mt.
Sinai, New York, NY
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN
| |
Collapse
|
43
|
Zhao PP, Xu LW, Sun T, Wu YY, Zhu XW, Zhang B, Cheng Z, Cai X, Liu YC, Zhao TT, Wu TT, Ma HY, Wang L, Zhang XW, Yang L, Zheng HF. Relationship between alcohol use, blood pressure and hypertension: an association study and a Mendelian randomisation study. J Epidemiol Community Health 2019; 73:796-801. [DOI: 10.1136/jech-2018-211185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 04/13/2019] [Accepted: 05/19/2019] [Indexed: 01/15/2023]
Abstract
BackgroundPast studies have found a strong relationship between alcohol drinking and human health.MethodsIn this study, we first tested the association of rs671 with alcohol use in 2349 participants in southeast China. We then evaluated the causal impact between alcohol use and cardiovascular traits through a Mendelian randomisation (MR) analysis.ResultsWe found strong evidence for the association of rs671 in the ALDH2 gene with alcohol drinking (p=6.08×10-47; ORadj G=4.50, 95% CI 3.67 to 5.52). We found that female G carriers of rs671 had a higher proportion of non-drinkers than male G carriers (88.01% vs 38.70%). In non-drinkers, the female G allele frequency was higher than the male G allele frequency (71.1% vs 55.2%). MR analysis suggested that alcohol use had a causal effect on blood pressure (increasing 9.46 mm Hg for systolic blood pressure (p=9.67×10-4) and 7.50 mm Hg for diastolic blood pressure (p=9.62×10-5)), and on hypertension in men (p=0.011; OR =1.19, 95% CI 1.04 to 1.36) and in pooled samples (p=0.013; OR =1.20, 95% CI 1.04 to 1.39), but not in women. We did not observe a causal effect of alcohol use on body mass index and lipid levels; further studies are needed to clarify the non-causal relationship.ConclusionsCompared to never-drinkers, current and previous alcohol use had a causal effect on blood pressure and hypertension in pooled samples and in men. These results reflect Chinese culture which does not encourage women to drink.
Collapse
|
44
|
Hartwell EE, Kranzler HR. Pharmacogenetics of alcohol use disorder treatments: an update. Expert Opin Drug Metab Toxicol 2019; 15:553-564. [PMID: 31162983 DOI: 10.1080/17425255.2019.1628218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Introduction: Alcohol use disorder (AUD) is highly prevalent; costly economically, socially, and interpersonally; and grossly undertreated. The low rate of utilization of medications with demonstrated (albeit modest) efficacy is particularly noteworthy. One approach to increasing the utility and safety of available medications is to use a precision medicine approach, which seeks to identify patients for whom specific medications are likely to be most efficacious and have the fewest adverse effects. Areas Covered: We review the literature on the pharmacogenetics of AUD treatment using both approved and off-label medications. We cover both laboratory studies and clinical trials, highlighting valuable mechanistic insights and underscoring the potential value of precision-based care for AUD. Expert Opinion: Pharmacotherapy can be a useful component of AUD treatment. Currently, the evidence regarding genetic predictors of medication efficacy is very limited. Thus, a precision medicine approach is not yet ready for widespread clinical implementation. Further research is needed to identify candidate genetic variants that moderate the response to both established and novel medications. The growing availability of large-scale, longitudinal datasets that enable the synthesis of genetic and electronic health record data provides important opportunities to develop this area of research.
Collapse
Affiliation(s)
- Emily E Hartwell
- a Mental Illness Research, Education and Clinical Center , Crescenz VAMC , Philadelphia , PA , USA.,b Center for Studies of Addiction, Department of Psychiatry , University of Pennsylvania Perelman School of Medicine , Philadelphia , PA , USA
| | - Henry R Kranzler
- a Mental Illness Research, Education and Clinical Center , Crescenz VAMC , Philadelphia , PA , USA.,b Center for Studies of Addiction, Department of Psychiatry , University of Pennsylvania Perelman School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
45
|
Legaz I, Pérez-Cárceles MD, de la Calle I, Arjona F, Roca M, Cejudo P, Luna A, Osuna E. Genetic susceptibility to nicotine and/or alcohol addiction: a systematic review. TOXIN REV 2019. [DOI: 10.1080/15569543.2019.1619085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Isabel Legaz
- University of Murcia - Espinardo Campus, Murcia, Spain
| | - M. D. Pérez-Cárceles
- Department of Legal and Forensic Medicine, Biomedical Research Institute (IMIB), Regional Campus of International Excellence “Campus Mare Nostrum”, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | | | - Miriam Roca
- University of Murcia - Espinardo Campus, Murcia, Spain
| | - Pablo Cejudo
- University of Murcia - Espinardo Campus, Murcia, Spain
| | - Aurelio Luna
- University of Murcia - Espinardo Campus, Murcia, Spain
| | - Eduardo Osuna
- University of Murcia - Espinardo Campus, Murcia, Spain
| |
Collapse
|
46
|
Hubacek JA, Pikhart H, Peasey A, Malyutina S, Pajak A, Tamosiunas A, Voevoda M, Holmes MV, Bobak M. The association between the FTO gene variant and alcohol consumption and binge and problem drinking in different gene-environment background: The HAPIEE study. Gene 2019; 707:30-35. [PMID: 31055022 DOI: 10.1016/j.gene.2019.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/07/2019] [Accepted: 05/01/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alcohol intake and tobacco smoking have significant negative health consequences and both are influenced by genetic predispositions. Some studies suggest that the FTO gene is associated with alcohol consumption. We investigated whether a tagging variant (rs17817449) within the FTO gene is associated with alcohol intake, problem drinking and smoking behaviour. METHODS We analysed data from 26,792 Caucasian adults (47.2% of males; mean age 58.9 (±7.3) years), examined through the prospective cohort HAPIEE study. The primary outcomes were daily alcohol consumption, binge drinking, problem drinking (CAGE score 2+) and smoking status in relation to tagging variants within the FTO and ADH1B genes. RESULTS We found no significant association of the FTO polymorphism with smoking status in either sex. The associations of the FTO polymorphism with drinking pattern were inconsistent and differed by gender. In men, GG homozygote carriers had lower odds of problem drinking (OR 0.85, 95% CI 0.75-0.96, p = 0.03). In women, the combination of the FTO/ADH1B GG/+A genotypes doubled the risk of binge drinking (OR 2.10, 95% CI 1.19-3.71, p < 0.05), and the risk was further increased among smoking women (OR 4.10, 95% CI 1.64-10.24, p = 0.008). CONCLUSIONS In this large population study, the FTO gene appeared associated with binge and problem drinking, and the associations were modified by sex, smoking status and the ADH1B polymorphism.
Collapse
Affiliation(s)
- Jaroslav A Hubacek
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Hynek Pikhart
- International Institute for Health and Society, Department of Epidemiology and Public Health, University College London, UK
| | - Anne Peasey
- International Institute for Health and Society, Department of Epidemiology and Public Health, University College London, UK
| | - Sofia Malyutina
- Research Institute of Internal and Preventive Medicine, Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
| | - Andrzej Pajak
- Department of Epidemiology and Population Studies, Institute of Public Health, Faculty of Health Care, Jagiellonian University Medical College, Krakow, Poland
| | - Abdonas Tamosiunas
- Department of Population Studies, Institute of Cardiology of Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mikhail Voevoda
- Research Institute of Internal and Preventive Medicine, Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
| | - Michael V Holmes
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, England, UK
| | - Martin Bobak
- International Institute for Health and Society, Department of Epidemiology and Public Health, University College London, UK
| |
Collapse
|
47
|
Smith RL, Salvatore JE, Aliev F, Neale Z, Barr P, Dick DM. Genes, Roommates, and Residence Halls: A Multidimensional Study of the Role of Peer Drinking on College Students' Alcohol Use. Alcohol Clin Exp Res 2019; 43:1254-1262. [PMID: 31034622 DOI: 10.1111/acer.14037] [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: 09/14/2018] [Accepted: 03/21/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Peer drinking is one of the most robust predictors of college students' alcohol use and can moderate students' genetic risk for alcohol use. Peer effect research generally suffers from 2 problems: selection into peer groups and relying more on perceptions of peer alcohol use than peers' self-report. The goal of the present study was to overcome those limitations by capitalizing on a genetically informed sample of randomly assigned college roommates to examine multiple dimensions of peer influence and the interplay between peer effects and genetic predisposition on alcohol use, in the form of polygenic scores. METHODS We used a subsample (n = 755) of participants from a university-wide, longitudinal study at a large, diverse, urban university. Participants reported their own alcohol use during fall and spring and their perceptions of college peers' alcohol use in spring. We matched individuals into their rooms and residence halls to create a composite score of peer-reported alcohol use for each of those levels. We examined multiple dimensions of peer influence and whether peer influence moderated genetic predisposition to predict college students' alcohol use using multilevel models to account for clustering at the room and residence hall level. RESULTS We found that polygenic scores (β = 0.12), perceptions of peer drinking (β = 0.37), and roommates' self-reported drinking (β = 0.10) predicted alcohol use (all ps < 0.001), while average alcohol use across residence hall did not (β = -0.01, p = 0.86). We found no evidence for interactions between peer influence and genome-wide polygenic scores for alcohol use. CONCLUSIONS Our findings underscore the importance of genetic predisposition on individual alcohol use and support the potentially causal nature of the association between peer influence and alcohol use.
Collapse
Affiliation(s)
- Rebecca L Smith
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia
| | - Jessica E Salvatore
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia.,Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, Virginia
| | - Fazil Aliev
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia.,Faculty of Business, Karabuk University, Karabuk, Turkey
| | - Zoe Neale
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia.,Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, Virginia
| | - Peter Barr
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia
| | | | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia.,College Behavioral and Emotional Health Institute, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
48
|
Mignogna KM, Bacanu SA, Riley BP, Wolen AR, Miles MF. Cross-species alcohol dependence-associated gene networks: Co-analysis of mouse brain gene expression and human genome-wide association data. PLoS One 2019; 14:e0202063. [PMID: 31017905 PMCID: PMC6481773 DOI: 10.1371/journal.pone.0202063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 04/07/2019] [Indexed: 01/06/2023] Open
Abstract
Genome-wide association studies on alcohol dependence, by themselves, have yet to account for the estimated heritability of the disorder and provide incomplete mechanistic understanding of this complex trait. Integrating brain ethanol-responsive gene expression networks from model organisms with human genetic data on alcohol dependence could aid in identifying dependence-associated genes and functional networks in which they are involved. This study used a modification of the Edge-Weighted Dense Module Searching for genome-wide association studies (EW-dmGWAS) approach to co-analyze whole-genome gene expression data from ethanol-exposed mouse brain tissue, human protein-protein interaction databases and alcohol dependence-related genome-wide association studies. Results revealed novel ethanol-responsive and alcohol dependence-associated gene networks in prefrontal cortex, nucleus accumbens, and ventral tegmental area. Three of these networks were overrepresented with genome-wide association signals from an independent dataset. These networks were significantly overrepresented for gene ontology categories involving several mechanisms, including actin filament-based activity, transcript regulation, Wnt and Syndecan-mediated signaling, and ubiquitination. Together, these studies provide novel insight for brain mechanisms contributing to alcohol dependence.
Collapse
Affiliation(s)
- Kristin M. Mignogna
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Center for Clinical & Translational Research, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Silviu A. Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Brien P. Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Aaron R. Wolen
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael F. Miles
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
49
|
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: 292] [Impact Index Per Article: 48.7] [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
|
50
|
Beeler E, Nobile ZL, Homanics GE. Paternal Preconception Every-Other-Day Ethanol Drinking Alters Behavior and Ethanol Consumption in Offspring. Brain Sci 2019; 9:brainsci9030056. [PMID: 30845665 PMCID: PMC6468863 DOI: 10.3390/brainsci9030056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 01/12/2023] Open
Abstract
Alcohol use disorder is a devastating disease with a complex etiology. Recent preclinical studies have revealed that paternal preconception chronic intermittent ethanol (EtOH) exposure via vaporized EtOH altered drinking behaviors and sensitivity to EtOH selectively in male offspring. In the current study, we used a voluntary oral route of paternal preconception EtOH exposure, i.e., intermittent every-other-day two-bottle choice drinking, and tested offspring for behavioral alterations. Fifteen EtOH drinking sires and 10 control sires were mated to EtOH naïve females to produce EtOH-sired and control-sired offspring. These offspring were tested using the elevated plus maze, open field, drinking in the dark, and unlimited access two-bottle choice assays. We found that paternal preconception every-other-day two-bottle choice drinking resulted in reduced EtOH consumption selectively in male offspring in the drinking in the dark assay compared to control-sired offspring. No differences were detected in either sex in the unlimited access two-bottle choice and elevated plus maze assays. Open field analysis revealed complex changes in basal behavior and EtOH-induced behaviors that were sex specific. We concluded that paternal preconception voluntary EtOH consumption has persistent effects that impact the next generation. This study adds to a growing appreciation that one’s behavioral response to EtOH and EtOH drinking behavior are impacted by EtOH exposure of the prior generation.
Collapse
Affiliation(s)
- Erik Beeler
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, 6068 Biomedical Science Tower-3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
| | - Zachary L Nobile
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA.
| | - Gregg E Homanics
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, 6068 Biomedical Science Tower-3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
- Center for Neuroscience, University of Pittsburgh School of Medicine, 6060 Biomedical Science Tower-3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 6060 Biomedical Science Tower-3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
- Department of Neurobiology, University of Pittsburgh School of Medicine, 6060 Biomedical Science Tower-3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
| |
Collapse
|