1
|
Wang C, Pu Q, Mo X, Han X, Wang F, Li W, Chen C, Xue Y, Xin J, Shen C, Du M, Wu D. A global overview of shared genetic architecture between smoking behaviors and major depressive disorder in European and East Asian ancestry. J Affect Disord 2025; 375:10-21. [PMID: 39842668 DOI: 10.1016/j.jad.2025.01.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/01/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND The co-occurrence of smoking behaviors and major depressive disorder (MDD) has been widely documented in populations. However, the underlying mechanism of this association remains unclear. METHODS Genome-wide association studies of smoking behaviors and MDD, combined with multi-omics datasets, were used to characterise genetic correlations, identify shared loci and genes, and explore underlying biological mechanisms. Mendelian randomization (MR) analyses were conducted to infer causal relationships between smoking behaviors and MDD. Druggability analyses were performed to identify potential drugs with both antidepressant and smoking cessation effects. RESULTS Extensive overall genetic correlations were found between smoking behaviors and MDD. Furthermore, eighteen local regions showed significant genetic correlations, which could be partly explained by gene co-expression patterns. We identified 24 shared loci and 120 genes, which were enriched in limbic system, GABAergic and dopaminergic neurons, as well as in synaptic pathways. Through integrating with tissue specific information, seven key genes (ANKK1, NEGR1, USP4, TCTA, SORCS5, SPPL3, and USP28) were pinpointed. Notably, druggability analyses supported ANKK1 as a potential drug target for the treatment of MDD and tobacco dependence. MR analyses suggested a bidirectional causal relationship between smoking initiation and MDD. Although findings in East Asian ancestry were limited, the shared locus (chr15:47613403-47,685,504) identified in European ancestry remained significant in East Asian ancestry. CONCLUSIONS Our findings suggest the extensive genetic overlap between smoking behaviors and MDD, support the role of limbic system and synapse involved in shared mechanisms, and implicate for prevention, intervention and treatment.
Collapse
Affiliation(s)
- Chao Wang
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuyi Pu
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoxiao Mo
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xu Han
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wen Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Changying Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yong Xue
- Department of Medical Laboratory, Huai'an No 3 People's Hospital, Huai'an, China
| | - Junyi Xin
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Mulong Du
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Dongmei Wu
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Liang Y, Ou J, Fu J, Wang Y, Li Y, Li J, Yi Y. Smoking, Genetic Susceptibility and Early Menopause: Unveiling Biological Mechanisms and Potential Therapy Targets. BJOG 2025; 132:625-637. [PMID: 39727065 DOI: 10.1111/1471-0528.18052] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To explore the association between smoking, genetic susceptibility and early menopause (EM) and clarify the potential mechanisms underlying this relationship. DESIGN An observational and Transcriptome-wide association analysis (TWAS) study. SETTING UK Biobank and public summary statistics. POPULATION 139 869 women with full baseline and menopause data, and no gynaecological surgery history. METHODS Adjusted modified Poisson regression models were developed to determine the smoking and genetic risk effects on EM. TWAS was used to identify gene expression between smoking and EM, with Mendelian randomisation (MR) to infer causality. Enrichment analysis explored regulatory networks of transcription factors, microRNAs and potential therapeutic targets. Small molecule drugs were predicted using drug-gene interaction analysis. MAIN OUTCOME MEASURES EM prevalence and common gene expression patterns. RESULTS Women with over 30 pack-years of smoking had about 1.5 times higher EM risk, with RRs of 1.39 (95%CI, 1.23-1.56), 1.45 (1.33-1.59) and 1.45 (1.36-1.55) in the low, intermediate and high genetic risk groups. TWAS identified hub genes such as IMMP2L, BMPR2 and HMGN1. MR confirmed daily cigarette consumption as a causal factor in early menopause. Several potential therapeutic targets (e.g., SP600125, INCB18424 and ruxolitinib) were identified. CONCLUSIONS Smoking reduction significantly lowered the risk of EM. Hub genes and therapeutic targets identified provided new avenues for mitigating harmful effects of smoking.
Collapse
Affiliation(s)
- Yuhang Liang
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Ou
- Department of Gyneacology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Fu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha, China
| | - Yijing Wang
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yanping Li
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha, China
| | - Jinchen Li
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yi
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Seah C, Karabacak M, Margetis K. Transcriptomic imputation identifies tissue-specific genes associated with cervical myelopathy. Spine J 2025; 25:588-596. [PMID: 39491753 DOI: 10.1016/j.spinee.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/25/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND CONTEXT Degenerative cervical myelopathy (DCM) is a progressive spinal condition that can lead to severe neurological dysfunction. Despite its degenerative pathophysiology, family history has shown to be a largely important factor in incidence and progression, suggesting that inherent genetic predisposition may play a role in pathophysiology. PURPOSE To determine the tissue-specific, functional genetic basis of hereditary predisposition to cervical myelopathy. STUDY DESIGN Retrospective case-control study using patient genetics and matched EHR from the Mount Sinai BioMe Biobank. METHODS In a large, diverse, urban biobank of 32,031 individuals, with 558 individuals with cervical myopathy, we applied transcriptomic imputation to identify genetically regulated gene expression signatures associated with DCM. We performed drug-repurposing analysis using the CMAP database to identify candidate therapeutic interventions to reverse the cervical myelopathy-associated gene signature. RESULTS We identified 16 genes significantly associated with DCM across 5 different tissues, suggesting tissue-specific manifestations of inherited genetic risk (upregulated: HES6, PI16, TMEM183A, BDH2, LINC00937, CLEC4D, USP43, SPATA1; downregulated: TTC12, CDK5, PAFAH1B2, RCSD1, KLHL29, PTPRG, RP11-620J15.3, C1RL). Drug repurposing identified 22 compounds with the potential to reverse the DCM-associated signature, suggesting points of therapeutic intervention. CONCLUSIONS The inherited genetic risk for cervical myelopathy is functionally associated with genes involved in tissue-specific nociceptive and proliferative processes. These signatures may be reversed by candidate therapeutics with nociceptive, calcium channel modulating, and antiproliferative effects. CLINICAL SIGNIFICANCE Understanding the genetic basis of DCM provides critical insights into the hereditary factors contributing to the disease, allowing for more personalized and targeted therapeutic approaches. The identification of candidate drugs through transcriptomic imputation and drug repurposing analysis offers potential new treatments that could significantly improve patient outcomes and quality of life by addressing the underlying genetic mechanisms of DCM.
Collapse
Affiliation(s)
- Carina Seah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA.
| | - Mert Karabacak
- Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA
| | | |
Collapse
|
4
|
Yang J, Wang H, Chen H, Hou H, Hu Q. The association of genetic polymorphisms within the dopaminergic system with nicotine dependence: A narrative review. Heliyon 2024; 10:e33158. [PMID: 39021905 PMCID: PMC11253068 DOI: 10.1016/j.heliyon.2024.e33158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Nicotine, the main compound in cigarettes, leads to smoking addiction. Nicotine acts on the limbic dopamine reward loop in the midbrain by binding to nicotinic acetylcholine receptors, promoting the release of dopamine, and resulting in a rewarding effect or satisfaction. This satisfaction is essential for continued and compulsive tobacco use, and therefore dopamine plays a crucial role in nicotine dependence. Numerous studies have identified genetic polymorphisms of dopaminergic pathways which may influence susceptibility to nicotine addiction. Dopamine levels are greatly influenced by synthesis, storage, release, degradation, and reuptake-related genes, including genes encoding tyrosine hydroxylase, dopamine decarboxylase, dopamine transporter, dopamine receptor, dopamine 3-hydroxylase, catechol-O-methyltransferase, and monoamine oxidase. In this paper, we review research progress on the effects of polymorphisms in the above genes on downstream smoking behavior and nicotine dependence, to offer a theoretical basis for the elucidation of the genetic mechanism underlying nicotine dependence and future personalized treatment for smoking cessation.
Collapse
Affiliation(s)
- Jingjing Yang
- China National Tobacco Quality Supervision & Test Center, Zhengzhou, 450001, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, 450001, China
- Beijing Life Science Academy, Beijing, 102209, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, 102209, China
| | - Hongjuan Wang
- China National Tobacco Quality Supervision & Test Center, Zhengzhou, 450001, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, 450001, China
- Beijing Life Science Academy, Beijing, 102209, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, 102209, China
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, Zhengzhou, 450001, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, 450001, China
- Beijing Life Science Academy, Beijing, 102209, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, 102209, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, Zhengzhou, 450001, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, 450001, China
- Beijing Life Science Academy, Beijing, 102209, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, 102209, China
| | - Qingyuan Hu
- China National Tobacco Quality Supervision & Test Center, Zhengzhou, 450001, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, 450001, China
- Beijing Life Science Academy, Beijing, 102209, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, 102209, China
| |
Collapse
|
5
|
Toikumo S, Jennings MV, Pham BK, Lee H, Mallard TT, Bianchi SB, Meredith JJ, Vilar-Ribó L, Xu H, Hatoum AS, Johnson EC, Pazdernik VK, Jinwala Z, Pakala SR, Leger BS, Niarchou M, Ehinmowo M, Jenkins GD, Batzler A, Pendegraft R, Palmer AA, Zhou H, Biernacka JM, Coombes BJ, Gelernter J, Xu K, Hancock DB, Cox NJ, Smoller JW, Davis LK, Justice AC, Kranzler HR, Kember RL, Sanchez-Roige S. Multi-ancestry meta-analysis of tobacco use disorder identifies 461 potential risk genes and reveals associations with multiple health outcomes. Nat Hum Behav 2024; 8:1177-1193. [PMID: 38632388 PMCID: PMC11199106 DOI: 10.1038/s41562-024-01851-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
Tobacco use disorder (TUD) is the most prevalent substance use disorder in the world. Genetic factors influence smoking behaviours and although strides have been made using genome-wide association studies to identify risk variants, most variants identified have been for nicotine consumption, rather than TUD. Here we leveraged four US biobanks to perform a multi-ancestral meta-analysis of TUD (derived via electronic health records) in 653,790 individuals (495,005 European, 114,420 African American and 44,365 Latin American) and data from UK Biobank (ncombined = 898,680). We identified 88 independent risk loci; integration with functional genomic tools uncovered 461 potential risk genes, primarily expressed in the brain. TUD was genetically correlated with smoking and psychiatric traits from traditionally ascertained cohorts, externalizing behaviours in children and hundreds of medical outcomes, including HIV infection, heart disease and pain. This work furthers our biological understanding of TUD and establishes electronic health records as a source of phenotypic information for studying the genetics of TUD.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Benjamin K Pham
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Hyunjoon Lee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Sevim B Bianchi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - John J Meredith
- 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
| | - Heng Xu
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Hatoum
- Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Emma C Johnson
- Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Zeal Jinwala
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shreya R Pakala
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Brittany S Leger
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Program in Biomedical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Maria Niarchou
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
| | | | - Greg D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Richard Pendegraft
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 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
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Ke Xu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | | | - Nancy J Cox
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Lea K Davis
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amy C Justice
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Public Health, New Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel L Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Blum K, Ashford JW, Kateb B, Sipple D, Braverman E, Dennen CA, Baron D, Badgaiyan R, Elman I, Cadet JL, Thanos PK, Hanna C, Bowirrat A, Modestino EJ, Yamamoto V, Gupta A, McLaughlin T, Makale M, Gold MS. Dopaminergic dysfunction: Role for genetic & epigenetic testing in the new psychiatry. J Neurol Sci 2023; 453:120809. [PMID: 37774561 DOI: 10.1016/j.jns.2023.120809] [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: 07/17/2021] [Revised: 08/02/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023]
Abstract
Reward Deficiency Syndrome (RDS), particularly linked to addictive disorders, costs billions of dollars globally and has resulted in over one million deaths in the United States (US). Illicit substance use has been steadily rising and in 2021 approximately 21.9% (61.2 million) of individuals living in the US aged 12 or older had used illicit drugs in the past year. However, only 1.5% (4.1 million) of these individuals had received any substance use treatment. This increase in use and failure to adequately treat or provide treatment to these individuals resulted in 106,699 overdose deaths in 2021 and increased in 2022. This article presents an alternative non-pharmaceutical treatment approach tied to gene-guided therapy, the subject of many decades of research. The cornerstone of this paradigm shift is the brain reward circuitry, brain stem physiology, and neurotransmitter deficits due to the effects of genetic and epigenetic insults on the interrelated cascade of neurotransmission and the net release of dopamine at the Ventral Tegmental Area -Nucleus Accumbens (VTA-NAc) reward site. The Genetic Addiction Risk Severity (GARS) test and pro-dopamine regulator nutraceutical KB220 were combined to induce "dopamine homeostasis" across the brain reward circuitry. This article aims to encourage four future actionable items: 1) the neurophysiologically accurate designation of, for example, "Hyperdopameism /Hyperdopameism" to replace the blaming nomenclature like alcoholism; 2) encouraging continued research into the nature of dysfunctional brainstem neurotransmitters across the brain reward circuitry; 3) early identification of people at risk for all RDS behaviors as a brain check (cognitive testing); 4) induction of dopamine homeostasis using "precision behavioral management" along with the coupling of GARS and precision Kb220 variants; 5) utilization of promising potential treatments include neuromodulating modalities such as Transmagnetic stimulation (TMS) and Deep Brain Stimulation(DBS), which target different areas of the neural circuitry involved in addiction and even neuroimmune agents like N-acetyl-cysteine.
Collapse
Affiliation(s)
- Kenneth Blum
- Division of Addiction Research & Education, Center for Exercise, Sports and Mental Health, Western University Health Sciences, Pomona, CA, USA; The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX, USA; Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel.
| | - J Wesson Ashford
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA; War Related Illness & Injury Study Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Babak Kateb
- Brain Mapping Foundation, Los Angeles, CA, USA; National Center for Nanobioelectronic, Los Angeles, CA, USA; Brain Technology and Innovation Park, Los Angeles, CA, USA
| | | | - Eric Braverman
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX, USA
| | - Catherine A Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA, USA
| | - David Baron
- Division of Addiction Research & Education, Center for Exercise, Sports and Mental Health, Western University Health Sciences, Pomona, CA, USA
| | - Rajendra Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX, USA; Long School of Medicine, University of Texas Medical Center, San Antonio, TX, USA
| | - Igor Elman
- Center for Pain and the Brain (PAIN Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Waltham, MA, USA; Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Bethesda, MD, USA
| | - Panayotis K Thanos
- Department of Psychology & Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Colin Hanna
- Department of Psychology & Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel
| | | | - Vicky Yamamoto
- Brain Mapping Foundation, Los Angeles, CA, USA; National Center for Nanobioelectronic, Los Angeles, CA, USA; Brain Technology and Innovation Park, Los Angeles, CA, USA; Society for Brain Mapping and Therapeutics, Los Angeles, CA, USA; USC-Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Thomas McLaughlin
- Division of Reward Deficiency Research, Reward Deficiency Syndrome Clinics of America, Austin, TX, USA
| | - Mlan Makale
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mark S Gold
- Department of Psychiatry, Washington College of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
Toikumo S, Jennings MV, Pham BK, Lee H, Mallard TT, Bianchi SB, Meredith JJ, Vilar-Ribó L, Xu H, Hatoum AS, Johnson EC, Pazdernik V, Jinwala Z, Pakala SR, Leger BS, Niarchou M, Ehinmowo M, Jenkins GD, Batzler A, Pendegraft R, Palmer AA, Zhou H, Biernacka JM, Coombes BJ, Gelernter J, Xu K, Hancock DB, Cox NJ, Smoller JW, Davis LK, Justice AC, Kranzler HR, Kember RL, Sanchez-Roige S. Multi-ancestry meta-analysis of tobacco use disorder prioritizes novel candidate risk genes and reveals associations with numerous health outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.27.23287713. [PMID: 37034728 PMCID: PMC10081388 DOI: 10.1101/2023.03.27.23287713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Tobacco use disorder (TUD) is the most prevalent substance use disorder in the world. Genetic factors influence smoking behaviors, and although strides have been made using genome-wide association studies (GWAS) to identify risk variants, the majority of variants identified have been for nicotine consumption, rather than TUD. We leveraged five biobanks to perform a multi-ancestral meta-analysis of TUD (derived via electronic health records, EHR) in 898,680 individuals (739,895 European, 114,420 African American, 44,365 Latin American). We identified 88 independent risk loci; integration with functional genomic tools uncovered 461 potential risk genes, primarily expressed in the brain. TUD was genetically correlated with smoking and psychiatric traits from traditionally ascertained cohorts, externalizing behaviors in children, and hundreds of medical outcomes, including HIV infection, heart disease, and pain. This work furthers our biological understanding of TUD and establishes EHR as a source of phenotypic information for studying the genetics of TUD.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mariela V Jennings
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Benjamin K Pham
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Hyunjoon Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Sevim B Bianchi
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - John J Meredith
- Department of Psychiatry, University of California San Diego, San Diego, 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
| | - Heng Xu
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Alexander S Hatoum
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Vanessa Pazdernik
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Zeal Jinwala
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shreya R Pakala
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Brittany S Leger
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
- Program in Biomedical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Maria Niarchou
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Greg D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Richard Pendegraft
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Ke Xu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Dana B Hancock
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Division, RTI International, Research Triangle Park, NC, USA
| | - Nancy J Cox
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amy C Justice
- Yale University School of Public Health, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel L Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
8
|
Holen B, Shadrin AA, Icick R, Filiz TT, Hindley G, Rødevand L, O’Connell KS, Hagen E, Frei O, Bahrami S, Cheng W, Parker N, Tesfaye M, Jahołkowski P, Karadag N, Dale AM, Djurovic S, Smeland OB, Andreassen OA. Genome-wide analyses reveal novel opioid use disorder loci and genetic overlap with schizophrenia, bipolar disorder, and major depression. Addict Biol 2023; 28:e13282. [PMID: 37252880 PMCID: PMC11896964 DOI: 10.1111/adb.13282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 06/01/2023]
Abstract
Opioid use disorder (OUD) and mental disorders are often comorbid, with increased morbidity and mortality. The causes underlying this relationship are poorly understood. Although these conditions are highly heritable, their shared genetic vulnerabilities remain unaccounted for. We applied the conditional/conjunctional false discovery rate (cond/conjFDR) approach to analyse summary statistics from independent genome wide association studies of OUD, schizophrenia (SCZ), bipolar disorder (BD) and major depression (MD) of European ancestry. Next, we characterized the identified shared loci using biological annotation resources. OUD data were obtained from the Million Veteran Program, Yale-Penn and Study of Addiction: Genetics and Environment (SAGE) (15 756 cases, 99 039 controls). SCZ (53 386 cases, 77 258 controls), BD (41 917 cases, 371 549 controls) and MD (170 756 cases, 329 443 controls) data were provided by the Psychiatric Genomics Consortium. We discovered genetic enrichment for OUD conditional on associations with SCZ, BD, MD and vice versa, indicating polygenic overlap with identification of 14 novel OUD loci at condFDR < 0.05 and 7 unique loci shared between OUD and SCZ (n = 2), BD (n = 2) and MD (n = 7) at conjFDR < 0.05 with concordant effect directions, in line with estimated positive genetic correlations. Two loci were novel for OUD, one for BD and one for MD. Three OUD risk loci were shared with more than one psychiatric disorder, at DRD2 on chromosome 11 (BD and MD), at FURIN on chromosome 15 (SCZ, BD and MD) and at the major histocompatibility complex region (SCZ and MD). Our findings provide new insights into the shared genetic architecture between OUD and SCZ, BD and MD, indicating a complex genetic relationship, suggesting overlapping neurobiological pathways.
Collapse
Affiliation(s)
- Børge Holen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Alexey A. Shadrin
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Romain Icick
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
- INSERM UMR-S1144, Paris University, F-75006, France
| | - Tahir T. Filiz
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Guy Hindley
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Linn Rødevand
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Kevin S. O’Connell
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Espen Hagen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Oleksandr Frei
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Shahram Bahrami
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Weiqiu Cheng
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Nadine Parker
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Markos Tesfaye
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Piotr Jahołkowski
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Naz Karadag
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Anders M. Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, United States of America
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA 92093, United States of America
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Olav B. Smeland
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| |
Collapse
|
9
|
Abstract
The current addiction crisis has destroyed a multitude of lives, leaving millions of fatalities worldwide in its wake. At the same time, various governmental agencies dedicated to solving this seemingly never-ending dilemma have not yet succeeded or delivered on their promises. We understand that addictive behavioral seeking is a multi-faceted neurobiological and spiritually complicated phenomenon. However, although the substitution replacement approach, especially to treat Opioid Use Disorder (OUD), has importance for harm reduction in the short term, it does not bring about a harm-free recovery or prevention. Instead, we propose a promising novel approach that uses genetic risk testing with induction of dopamine homeostasis and an objective Brain Health Check during youth. Our model involves a six-hit approach known as the "Reward Dysregulation Syndrome Solution System," which can identify addiction risk and target the root cause of addiction, dopamine dysregulation. While we applaud all past sophisticated neurogenetic and neuropharmacological research, our opinion is that in the long term, addiction scientists and clinicians might characterize preaddiction using tests; for example, administering the validated RDSQuestionarre29, genetic risk assessment, a modified brain health check, or diagnostic framing of mild to moderate Substance Use Disorder (SUD). The preaddiction concept could incentivize the development of interventions to prevent addiction from developing in the first place and target and treat neurotransmitter imbalances and other early indications of addiction. WC 222.
Collapse
|
10
|
Blum K, McLaughlin T, Bowirrat A, Modestino EJ, Baron D, Gomez LL, Ceccanti M, Braverman ER, Thanos PK, Cadet JL, Elman I, Badgaiyan RD, Jalali R, Green R, Simpatico TA, Gupta A, Gold MS. Reward Deficiency Syndrome (RDS) Surprisingly Is Evolutionary and Found Everywhere: Is It "Blowin' in the Wind"? J Pers Med 2022; 12:jpm12020321. [PMID: 35207809 PMCID: PMC8875142 DOI: 10.3390/jpm12020321] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/04/2022] Open
Abstract
Reward Deficiency Syndrome (RDS) encompasses many mental health disorders, including a wide range of addictions and compulsive and impulsive behaviors. Described as an octopus of behavioral dysfunction, RDS refers to abnormal behavior caused by a breakdown of the cascade of reward in neurotransmission due to genetic and epigenetic influences. The resultant reward neurotransmission deficiencies interfere with the pleasure derived from satisfying powerful human physiological drives. Epigenetic repair may be possible with precision gene-guided therapy using formulations of KB220, a nutraceutical that has demonstrated pro-dopamine regulatory function in animal and human neuroimaging and clinical trials. Recently, large GWAS studies have revealed a significant dopaminergic gene risk polymorphic allele overlap between depressed and schizophrenic cohorts. A large volume of literature has also identified ADHD, PTSD, and spectrum disorders as having the known neurogenetic and psychological underpinnings of RDS. The hypothesis is that the true phenotype is RDS, and behavioral disorders are endophenotypes. Is it logical to wonder if RDS exists everywhere? Although complex, “the answer is blowin’ in the wind,” and rather than intangible, RDS may be foundational in species evolution and survival, with an array of many neurotransmitters and polymorphic loci influencing behavioral functionality.
Collapse
Affiliation(s)
- Kenneth Blum
- Division of Addiction Research & Education, Center for Psychiatry, Medicine, & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA;
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, (Ivitalize, Inc.), Austin, TX 78701, USA; (L.L.G.); (E.R.B.); (R.J.); (R.G.)
- Department of Psychiatry, University of Vermont, Burlington, VT 05405, USA;
- Department of Psychiatry, Wright University Boonshoff School of Medicine, Dayton, OH 45324, USA
- Correspondence: ; Tel.: +1-619-890-2167
| | | | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | | | - David Baron
- Division of Addiction Research & Education, Center for Psychiatry, Medicine, & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Luis Llanos Gomez
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, (Ivitalize, Inc.), Austin, TX 78701, USA; (L.L.G.); (E.R.B.); (R.J.); (R.G.)
| | - Mauro Ceccanti
- Alcohol Addiction Program, Latium Region Referral Center, Sapienza University of Rome, 00185 Roma, Italy;
| | - Eric R. Braverman
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, (Ivitalize, Inc.), Austin, TX 78701, USA; (L.L.G.); (E.R.B.); (R.J.); (R.G.)
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14203, USA;
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA;
| | - Igor Elman
- Center for Pain and the Brain (PAIN Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA;
- Cambridge Health Alliance, Harvard Medical School, Cambridge, MA 02139, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, Long School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA;
- Department of Psychiatry, MT. Sinai School of Medicine, New York, NY 10003, USA
| | - Rehan Jalali
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, (Ivitalize, Inc.), Austin, TX 78701, USA; (L.L.G.); (E.R.B.); (R.J.); (R.G.)
| | - Richard Green
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, (Ivitalize, Inc.), Austin, TX 78701, USA; (L.L.G.); (E.R.B.); (R.J.); (R.G.)
| | | | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA;
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
11
|
Dwyer DS. Protein Receptors Evolved from Homologous Cohesion Modules That Self-Associated and Are Encoded by Interactive Networked Genes. Life (Basel) 2021; 11:life11121335. [PMID: 34947866 PMCID: PMC8707797 DOI: 10.3390/life11121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Previously, it was proposed that protein receptors evolved from self-binding peptides that were encoded by self-interacting gene segments (inverted repeats) widely dispersed in the genome. In addition, self-association of the peptides was thought to be mediated by regions of amino acid sequence similarity. To extend these ideas, special features of receptors have been explored, such as their degree of homology to other proteins, and the arrangement of their genes for clues about their evolutionary origins and dynamics in the genome. As predicted, BLASTP searches for homologous proteins detected a greater number of unique hits for queries with receptor sequences than for sequences of randomly-selected, non-receptor proteins. This suggested that the building blocks (cohesion modules) for receptors were duplicated, dispersed, and maintained in the genome, due to structure/function relationships discussed here. Furthermore, the genes coding for a representative panel of receptors participated in a larger number of gene-gene interactions than for randomly-selected genes. This could conceivably reflect a greater evolutionary conservation of the receptor genes, with their more extensive integration into networks along with inherent properties of the genes themselves. In support of the latter possibility, some receptor genes were located in active areas of adaptive gene relocation/amalgamation to form functional blocks of related genes. It is suggested that adaptive relocation might allow for their joint regulation by common promoters and enhancers, and affect local chromatin structural domains to facilitate or repress gene expression. Speculation is included about the nature of the coordinated communication between receptors and the genes that encode them.
Collapse
Affiliation(s)
- Donard S Dwyer
- Departments of Psychiatry and Behavioral Medicine and Pharmacology, Toxicology and Neuroscience, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71130, USA
| |
Collapse
|
12
|
Genomic and Personalized Medicine Approaches for Substance Use Disorders (SUDs) Looking at Genome-Wide Association Studies. Biomedicines 2021; 9:biomedicines9121799. [PMID: 34944615 PMCID: PMC8698472 DOI: 10.3390/biomedicines9121799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
Drug addiction, or substance use disorder (SUD), is a chronic, relapsing disorder in which compulsive drug-seeking and drug-taking behaviour persist despite serious negative consequences. Drug abuse represents a problem that deserves great attention from a social point of view, and focuses on the importance of genetic studies to help in understanding the genetic basis of addiction and its medical treatment. Despite the complexity of drug addiction disorders, and the high number of environmental variables playing a role in the onset, recurrence, and duration of the symptoms, several studies have highlighted the non-negligible role of genetics, as demonstrated by heritability and genome-wide association studies. A correlation between the relative risk of addiction to specific substances and heritability has been recently observed, suggesting that neurobiological mechanisms may be, at least in part, inherited. All these observations point towards a scenario where the core neurobiological factors of addiction, involving the reward system, impulsivity, compulsivity, stress, and anxiety response, are transmitted, and therefore, genes and mutations underlying their variation might be detected. In the last few years, the development of new and more efficient sequencing technologies has paved the way for large-scale studies in searching for genetic and epigenetic factors affecting drug addiction disorders and their treatments. These studies have been crucial to pinpoint single nucleotide polymorphisms (SNPs) in genes that affect the reaction to medical treatments. This is critically important to identify pharmacogenomic approaches for substance use disorder, such as OPRM1 SNPs and methadone required doses for maintenance treatment (MMT). Nevertheless, despite the promising results obtained by genome-wide association and pharmacogenomic studies, specific studies related to population genetics diversity are lacking, undermining the overall applicability of the preliminary findings, and thus potentially affecting the portability and the accuracy of the genetic studies. In this review, focusing on cannabis, cocaine and heroin use, we report the state-of-the-art genomics and pharmacogenomics of SUDs, and the possible future perspectives related to medical treatment response in people that ask for assistance in solving drug-related problems.
Collapse
|
13
|
Ye Z, Mo C, Liu S, Hatch KS, Gao S, Ma Y, Hong LE, Thompson PM, Jahanshad N, Acheson A, Garavan H, Shen L, Nichols TE, Kochunov P, Chen S, Ma T. White Matter Integrity and Nicotine Dependence: Evaluating Vertical and Horizontal Pleiotropy. Front Neurosci 2021; 15:738037. [PMID: 34720862 PMCID: PMC8551454 DOI: 10.3389/fnins.2021.738037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/01/2021] [Indexed: 01/26/2023] Open
Abstract
Tobacco smoking is an addictive behavior that supports nicotine dependence and is an independent risk factor for cancer and other illnesses. Its neurogenetic mechanisms are not fully understood but may act through alterations in the cerebral white matter (WM). We hypothesized that the vertical pleiotropic pathways, where genetic variants influence a trait that in turn influences another trait, link genetic factors, integrity of cerebral WM, and nicotine addiction. We tested this hypothesis using individual genetic factors, WM integrity measured by fractional anisotropy (FA), and nicotine dependence-related smoking phenotypes, including smoking status (SS) and cigarettes per day (CPDs), in a large epidemiological sample collected by the UK Biobank. We performed a genome-wide association study (GWAS) to identify previously reported loci associated with smoking behavior. Smoking was found to be associated with reduced WM integrity in multiple brain regions. We then evaluated two competing vertical pathways: Genes → WM integrity → Smoking versus Genes → Smoking → WM integrity and a horizontal pleiotropy pathway where genetic factors independently affect both smoking and WM integrity. The causal pathway analysis identified 272 pleiotropic single-nucleotide polymorphisms (SNPs) whose effects on SS were mediated by FA, as well as 22 pleiotropic SNPs whose effects on FA were mediated by CPD. These SNPs were mainly located in important susceptibility genes for smoking-induced diseases NCAM1 and IREB2. Our findings revealed the role of cerebral WM in the maintenance of the complex addiction and provided potential genetic targets for future research in examining how changes in WM integrity contribute to the nicotine effects on the brain.
Collapse
Affiliation(s)
- Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Chen Mo
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Song Liu
- School of Computer Science and Technology, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Kathryn S Hatch
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Yizhou Ma
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Ashley Acheson
- Department of Psychiatry and Behavioral Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Hugh Garavan
- Department of Psychiatry, The University of Vermont, Burlington, VT, United States
| | - Li Shen
- Department of Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas E Nichols
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, College Park, MD, United States
| |
Collapse
|
14
|
An association study in the Taiwan Biobank elicits three novel candidates for cognitive aging in old adults: NCAM1, TTC12 and ZBTB20. Aging (Albany NY) 2021; 13:18769-18788. [PMID: 34285142 PMCID: PMC8351692 DOI: 10.18632/aging.203321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/08/2021] [Indexed: 01/11/2023]
Abstract
The dopamine receptor-related loci have been suggested to be associated with cognitive functions and neurodegenerative diseases. It is unknown whether genetic variants such as single nucleotide polymorphisms (SNPs) in the dopamine receptor-related loci could contribute to cognitive aging independently as well as by virtue of complicated interplays in the elder population. To assess whether SNPs in the dopamine receptor-related loci are associated with cognitive aging in the elder population, we evaluated SNPs in the DRD1, NCAM1-TTC12-ANKK1-DRD2, DRD3-LOC107986115-ZNF80-TIGIT-MIR568-ZBTB20, DRD4, and DRD5-SLC2A9 loci from 25,195 older Taiwanese individuals from the Taiwan Biobank. Mini-Mental State Examination (MMSE) was scrutinized for all participants, where MMSE scores were employed to evaluate cognitive functions. From our analysis, we identified three novel genes for cognitive aging that have not previously been reported: ZBTB20 on chromosome 3 and NCAM1 and TTC12 on chromosome 11. NCAM1 and ZBTB20 are strong candidates for having a role in cognitive aging with mutations in ZBTB20 resulting in intellectual disability, and NCAM1 previously found to be associated with associative memory in humans. Additionally, we found the effects of interplays between physical activity and these three novel genes. Our study suggests that genetic variants in the dopamine receptor-related loci may influence cognitive aging individually and by means of gene-physical activity interactions.
Collapse
|
15
|
Aliasghari F, Nazm SA, Yasari S, Mahdavi R, Bonyadi M. Associations of the ANKK1 and DRD2 gene polymorphisms with overweight, obesity and hedonic hunger among women from the Northwest of Iran. Eat Weight Disord 2021; 26:305-312. [PMID: 32020513 DOI: 10.1007/s40519-020-00851-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Pleasure from palatable foods can stimulate hedonic eating and, therefore, might be a major culprit for obesity. Dopamine receptor polymorphisms, especially variants in the genes regulating the D2 receptor, including ANKK1 and DRD2, are the prime candidates for assessing the individual differences in hedonic eating. This study was carried out to investigate the possible associations of the T (rs1800497) and Del (rs1799732) alleles with body mass index (BMI) and hedonic hunger among Iranian Azeri women. METHODS A total of 372 healthy overweight/obese subjects (BMI ≥ 25 kg/m2) and 159 normal weight individuals (BMI 18.5-24.9 kg/m2) were genotyped for the polymorphisms of ANNK1 and DRD2 genes using PCR-RFLP. BMI and hedonic hunger were also evaluated. RESULTS Three hundred and sixty-three (68.36%), 152 (28.63%), and 16 (3.01%) of the participants had CC, CT, and TT genotypes for ANNK1 gene, respectively. Of 515 samples genotyped for DRD2 gene, 315 (60.51%), 173 (33.59%), and 27 (5.24%) had Ins/Ins, Ins/Del, and Del/Del genotypes, respectively. The genotype and genotype frequencies were significantly different between the groups (p = 0.04). Significant differences were observed between the T+ genotype (TT + TC) and the T- genotype (CC) regarding the BMI and hedonic hunger scores (p < 0.05). In addition, Del+ group (Del/Del + Ins/Del) had higher BMI and hedonic hunger scores compared to Del- group (Ins/Ins) (p < 0.05). CONCLUSIONS Our findings showed that the frequencies of T and Del alleles were greater in the overweight/obese individuals. Also, the polymorphism of ANKK1 (rs1800497) and polymorphism of the DRD2 gene (rs1799732) showed significant associations with BMI and hedonic hunger. LEVEL OF EVIDENCE Level III, case-control study.
Collapse
Affiliation(s)
- Fereshteh Aliasghari
- Nutrition Research Center, Student Research Committee, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba A Nazm
- Faculty of Natural Sciences, Center of Excellence for Biodiversity, University of Tabriz, Tabriz, Iran
| | - Sepideh Yasari
- Faculty of Natural Sciences, Center of Excellence for Biodiversity, University of Tabriz, Tabriz, Iran
| | - Reza Mahdavi
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Morteza Bonyadi
- Faculty of Natural Sciences, Center of Excellence for Biodiversity, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
16
|
Boswell RG, Potenza MN, Grilo CM. The Neurobiology of Binge-eating Disorder Compared with Obesity: Implications for Differential Therapeutics. Clin Ther 2021; 43:50-69. [PMID: 33257092 PMCID: PMC7902428 DOI: 10.1016/j.clinthera.2020.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Emerging work indicates divergence in the neurobiologies of binge-eating disorder (BED) and obesity despite their frequent co-occurrence. This review highlights specific distinguishing aspects of BED, including elevated impulsivity and compulsivity possibly involving the mesocorticolimbic dopamine system, and discusses implications for differential therapeutics for BED. METHODS This narrative review describes epidemiologic, clinical, genetic, and preclinical differences between BED and obesity. Subsequently, this review discusses human neuroimaging work reporting differences in executive functioning, reward processing, and emotion reactivity in BED compared with obesity. Finally, on the basis of the neurobiology of BED, this review identifies existing and new therapeutic agents that may be most promising given their specific targets based on putative mechanisms of action relevant specifically to BED. FINDINGS BED is characterized by elevated impulsivity and compulsivity compared with obesity, which is reflected in divergent neurobiological characteristics and effective pharmacotherapies. Therapeutic agents that influence both reward and executive function systems may be especially effective for BED. IMPLICATIONS Greater attention to impulsivity/compulsivity-related, reward-related, and emotion reactivity-related processes may enhance conceptualization and treatment approaches for patients with BED. Consideration of these distinguishing characteristics and processes could have implications for more targeted pharmacologic treatment research and interventions.
Collapse
Affiliation(s)
- Rebecca G Boswell
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA.
| | - Marc N Potenza
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA; Connecticut Mental Health Center, New Haven, CT, USA; Connecticut Council on Problem Gambling, Wethersfield, CT, USA; Yale School of Medicine, Child Study Center, New Haven, CT, USA; Yale University, Department of Neuroscience, New Haven, CT, USA
| | - Carlos M Grilo
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA; Yale University, Department of Psychology, New Haven, CT, USA
| |
Collapse
|
17
|
Liu Q, Xu Y, Mao Y, Ma Y, Wang M, Han H, Cui W, Yuan W, Payne TJ, Xu Y, Li MD, Yang Z. Genetic and Epigenetic Analysis Revealing Variants in the NCAM1-TTC12-ANKK1-DRD2 Cluster Associated Significantly With Nicotine Dependence in Chinese Han Smokers. Nicotine Tob Res 2020; 22:1301-1309. [PMID: 31867628 DOI: 10.1093/ntr/ntz240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 12/17/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUNDS Although studies have demonstrated that the NCAM1-TTC12-ANKK1-DRD2 gene cluster plays essential roles in addictions in subjects of European and African origin, study of Chinese Han subjects is limited. Further, the underlying biological mechanisms of detected associations are largely unknown. METHODS Sixty-four single-nucleotide polymorphisms (SNPs) in this cluster were analyzed for association with Fagerstrőm Test for Nicotine Dependence score (FTND) and cigarettes per day (CPD) in male Chinese Han smokers (N = 2616). Next-generation bisulfite sequencing was used to discover smoking-associated differentially methylated regions (DMRs). Both cis-eQTL and cis-mQTL analyses were applied to assess the cis-regulatory effects of these risk SNPs. RESULTS Association analysis revealed that rs4648317 was significantly associated with FTND and CPD (p = .00018; p = .00072). Moreover, 14 additional SNPs were marginally significantly associated with FTND or CPD (p = .05-.01). Haplotype-based association analysis showed that one haplotype in DRD2, C-T-A-G, formed by rs4245148, rs4581480, rs4648317, and rs11214613, was significantly associated with CPD (p = .0005) and marginally associated with FTND (p = .003). Further, we identified four significant smoking-associated DMRs, three of which are located in the DRD2/ANKK1 region (p = .0012-.00005). Finally, we found five significant CpG-SNP pairs (p = 7.9 × 10-9-6.6 × 10-6) formed by risk SNPs rs4648317, rs11604671, and rs2734849 and three methylation loci. CONCLUSIONS We found two missense variants (rs11604671; rs2734849) and an intronic variant (rs4648317) with significant effects on ND and further explored their mechanisms of action through expression and methylation analysis. We found the majority of smoking-related DMRs are located in the ANKK1/DRD2 region, indicating a likely causative relation between non-synonymous SNPs and DMRs. IMPLICATIONS This study shows that there exist significant association of variants and haplotypes in ANKK1/DRD2 region with ND in Chinese male smokers. Further, this study also shows that DNA methylation plays an important role in mediating such associations.
Collapse
Affiliation(s)
- Qiang Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Mao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunlong Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Maiqiu Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haijun Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenyan Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenji Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Thomas J Payne
- ACT Center for Tobacco Treatment, Education and Research, Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS
| | - Yizhou Xu
- The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Xu K, Li B, McGinnis KA, Vickers-Smith R, Dao C, Sun N, Kember RL, Zhou H, Becker WC, Gelernter J, Kranzler HR, Zhao H, Justice AC. Genome-wide association study of smoking trajectory and meta-analysis of smoking status in 842,000 individuals. Nat Commun 2020; 11:5302. [PMID: 33082346 PMCID: PMC7598939 DOI: 10.1038/s41467-020-18489-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
Here we report a large genome-wide association study (GWAS) for longitudinal smoking phenotypes in 286,118 individuals from the Million Veteran Program (MVP) where we identified 18 loci for smoking trajectory of current versus never in European Americans, one locus in African Americans, and one in Hispanic Americans. Functional annotations prioritized several dozen genes where significant loci co-localized with either expression quantitative trait loci or chromatin interactions. The smoking trajectories were genetically correlated with 209 complex traits, for 33 of which smoking was either a causal or a consequential factor. We also performed European-ancestry meta-analyses for smoking status in the MVP and GWAS & Sequencing Consortium of Alcohol and Nicotine use (GSCAN) (Ntotal = 842,717) and identified 99 loci for smoking initiation and 13 loci for smoking cessation. Overall, this large GWAS of longitudinal smoking phenotype in multiple populations, combined with a meta-GWAS for smoking status, adds new insights into the genetic vulnerability for smoking behavior.
Collapse
Affiliation(s)
- Ke Xu
- Yale School of Medicine, New Haven, CT, 06511, USA
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Boyang Li
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | | | | | - Cecilia Dao
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - Ning Sun
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - Rachel L Kember
- 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
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - William C Becker
- Yale School of Medicine, New Haven, CT, 06511, USA
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Joel Gelernter
- Yale School of Medicine, New Haven, CT, 06511, USA
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Henry R Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Hongyu Zhao
- Yale School of Medicine, New Haven, CT, 06511, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - Amy C Justice
- Yale School of Medicine, New Haven, CT, 06511, USA.
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
19
|
Kaminskaite M, Jokubka R, Janaviciute J, Lelyte I, Sinkariova L, Pranckeviciene A, Borutaite V, Bunevicius A. Epistatic effect of Ankyrin repeat and kinase domain containing 1 - Dopamine receptor D2 and catechol-o-methyltransferase single nucleotide polymorphisms on the risk for hazardous use of alcohol in Lithuanian population. Gene 2020; 765:145107. [PMID: 32889058 DOI: 10.1016/j.gene.2020.145107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
AIM The Lithuanian population has outstanding rates of alcohol consumption and alcohol related mortality. Alteration of brain dopaminergic system play a role in the risk for addiction disorders. We evaluated the association of one single nucleotide polymorphism rs1800497 in the Ankyrin Repeat and Kinase Domain Containing 1 - Dopamine Receptor D2 complex (ANKK1-DRD2) and a catechol-o-methyltransferase (COMT) rs4680 single nucleotide polymorphism with the risk for alcohol use disorder and impulsiveness in Lithuanian population. Both genetic polymorphisms are known to alter brain dopaminergic activity, thus we also investigated the possible interaction effect of these polymorphisms. METHODS The study included 329 participants recruited from the local community. Hazardous alcohol use was evaluated using the Alcohol Use Disorder Identification Test (AUDIT). Impulsiveness was measured using the Barratt Impulsiveness Scale - 11 (BIS-11). Between group differences of AUDIT and BIS-11 scores were examined stratified by genetic polymorphisms and their combinations. The independent effect of each polymorphism and their interaction for hazardous alcohol use were evaluated using adjusted logistic regression analyses. RESULTS The ANKK1-DRD2 rs1800497 polymorphism was associated with total AUDIT score, but not with the hazardous use of alcohol, as indicated by the AUDIT test cut-off of 8. The COMT rs4680 GG genotype was associated with the hazardous use of alcohol (adjusted OR = 2.094, p = 0.029), but this association was not statistically significant after adjustment for multiple comparisons. Presence of both COMT rs4680 and ANKK1-DRD2 rs1800497 GGxCT/TT polymorphisms was associated with significantly increased risk for hazardous use of alcohol (adjusted OR = 5.016, p = 0.005). The COMT rs4680 and ANKK1-DRD2 rs1800497 genetic polymorphisms, and their combination were not associated with impulsiveness. CONCLUSIONS Our study demonstrated that the interaction of COMT rs4680 and ANKK1-DRD2 rs1800497 genetic polymorphisms is associated with a hazardous use of alcohol.
Collapse
Affiliation(s)
- Migle Kaminskaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Ramunas Jokubka
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Inesa Lelyte
- Department of Psychology, Vytautas Magnus University, Kaunas, Lithuania
| | - Liuda Sinkariova
- Department of Psychology, Vytautas Magnus University, Kaunas, Lithuania
| | - Aiste Pranckeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Adomas Bunevicius
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
20
|
Koeneke A, Ponce G, Troya-Balseca J, Palomo T, Hoenicka J. Ankyrin Repeat and Kinase Domain Containing 1 Gene, and Addiction Vulnerability. Int J Mol Sci 2020; 21:ijms21072516. [PMID: 32260442 PMCID: PMC7177674 DOI: 10.3390/ijms21072516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 01/13/2023] Open
Abstract
The TaqIA single nucleotide variant (SNV) has been tested for association with addictions in a huge number of studies. TaqIA is located in the ankyrin repeat and kinase domain containing 1 gene (ANKK1) that codes for a receptor interacting protein kinase. ANKK1 maps on the NTAD cluster along with the dopamine receptor D2 (DRD2), the tetratricopeptide repeat domain 12 (TTC12) and the neural cell adhesion molecule 1 (NCAM1) genes. The four genes have been associated with addictions, although TTC12 and ANKK1 showed the strongest associations. In silico and in vitro studies revealed that ANKK1 is functionally related to the dopaminergic system, in particular with DRD2. In antisocial alcoholism, epistasis between ANKK1 TaqIA and DRD2 C957T SNVs has been described. This clinical finding has been supported by the study of ANKK1 expression in peripheral blood mononuclear cells of alcoholic patients and controls. Regarding the ANKK1 protein, there is direct evidence of its location in adult and developing central nervous system. Together, these findings of the ANKK1 gene and its protein suggest that the TaqIA SNV is a marker of brain differences, both in structure and in dopaminergic function, that increase individual risk to addiction development.
Collapse
Affiliation(s)
- Alejandra Koeneke
- Departamento de Psicología, Facultad de Ciencias Biomédicas, Universidad Europea Madrid, Villaviciosa de Odón, 28670 Madrid, Spain;
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Guillermo Ponce
- Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Av. de Córdoba s/n, 28041 Madrid, Spain;
| | - Johanna Troya-Balseca
- Laboratory of Neurogenetics and Molecular Medicine - IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Tomás Palomo
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Janet Hoenicka
- Laboratory of Neurogenetics and Molecular Medicine - IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-936009751 (ext. 77833)
| |
Collapse
|
21
|
Grzywacz A, Suchanecka A, Chmielowiec J, Chmielowiec K, Szumilas K, Masiak J, Balwicki Ł, Michałowska-Sawczyn M, Trybek G. Personality Traits or Genetic Determinants-Which Strongly Influences E-Cigarette Users? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17010365. [PMID: 31948125 PMCID: PMC6981659 DOI: 10.3390/ijerph17010365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/21/2022]
Abstract
Presently, a growing popularity of electronic cigarettes may be observed. Used as a means of obtaining nicotine they allow to substitute traditional cigarettes. The origins of substance use disorders are conditioned by dopaminergic signaling which influences motivational processes being elementary factors conditioning the process of learning and exhibiting goal-directed behaviors. The study concentrated on analysis of three polymorphisms located in the dopamine receptor 2 (DRD2) gene—rs1076560, rs1799732 and rs1079597 using the PCR method, personality traits determined with the Big Five Questionnaire, and anxiety measured with the State Trait Anxiety Inventory. The study was conducted on a group of 394 volunteers, consisting e-cigarette users (n = 144) and controls (n = 250). Compared to the controls the case group subjects achieved significantly higher scores in regard to the STAI state and the trait scale, as well as the NEO-FFI Neuroticism and Openness scale. Likewise, in the case of the STAI state for DRD2 rs1076560 significant differences were found. Furthermore, while comparing the groups (e-cigarette users vs. controls) we noticed interactions for the NEO FFI Neuroticism and DRD2 rs1076560. The same was observed in the case of interactions significance while comparing groups (e-cigarette users vs. controls) for the STAI trait/scale and DRD2 rs1799732. Findings from this study demonstrate that psychological factors and genetic determinants should be analyzed simultaneously and comprehensively while considering groups of addicted patients. Since the use, and rapid increase in popularity, of electronic cigarettes has implications for public health, e-cigarette users should be studied holistically, especially younger groups of addicted and experimenting users.
Collapse
Affiliation(s)
- Anna Grzywacz
- Independent Laboratory of Health Promotion of the Pomeranian Medical University in Szczecin, 11 Chlapowskiego St., 70-204 Szczecin, Poland;
- Correspondence:
| | - Aleksandra Suchanecka
- Independent Laboratory of Health Promotion of the Pomeranian Medical University in Szczecin, 11 Chlapowskiego St., 70-204 Szczecin, Poland;
| | - Jolanta Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (J.C.); (K.C.)
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (J.C.); (K.C.)
| | - Kamila Szumilas
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp.72, 70-111 Szczecin, Poland;
| | - Jolanta Masiak
- Neurophysiological Independent Unit, Department of Psychiatry, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Łukasz Balwicki
- Department of Public Health and Social Medicine, Medical University of Gdansk, 42A Zwyciestwa St., 80-210 Gdansk, Poland;
| | | | - Grzegorz Trybek
- Department of Oral Surgery, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. St., 70-111 Szczecin, Poland;
| |
Collapse
|
22
|
Liu S, Rao S, Xu Y, Li J, Huang H, Zhang X, Fu H, Wang Q, Cao H, Baranova A, Jin C, Zhang F. Identifying common genome-wide risk genes for major psychiatric traits. Hum Genet 2019; 139:185-198. [DOI: 10.1007/s00439-019-02096-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/24/2019] [Indexed: 10/25/2022]
|
23
|
Evangelou E, Gao H, Chu C, Ntritsos G, Blakeley P, Butts AR, Pazoki R, Suzuki H, Koskeridis F, Yiorkas AM, Karaman I, Elliott J, Luo Q, Aeschbacher S, Bartz TM, Baumeister SE, Braund PS, Brown MR, Brody JA, Clarke TK, Dimou N, Faul JD, Homuth G, Jackson AU, Kentistou KA, Joshi PK, Lemaitre RN, Lind PA, Lyytikäinen LP, Mangino M, Milaneschi Y, Nelson CP, Nolte IM, Perälä MM, Polasek O, Porteous D, Ratliff SM, Smith JA, Stančáková A, Teumer A, Tuominen S, Thériault S, Vangipurapu J, Whitfield JB, Wood A, Yao J, Yu B, Zhao W, Arking DE, Auvinen J, Liu C, Männikkö M, Risch L, Rotter JI, Snieder H, Veijola J, Blakemore AI, Boehnke M, Campbell H, Conen D, Eriksson JG, Grabe HJ, Guo X, van der Harst P, Hartman CA, Hayward C, Heath AC, Jarvelin MR, Kähönen M, Kardia SLR, Kühne M, Kuusisto J, Laakso M, Lahti J, Lehtimäki T, McIntosh AM, Mohlke KL, Morrison AC, Martin NG, Oldehinkel AJ, Penninx BWJH, Psaty BM, Raitakari OT, Rudan I, Samani NJ, Scott LJ, Spector TD, Verweij N, Weir DR, Wilson JF, Levy D, Tzoulaki I, Bell JD, Matthews PM, Rothenfluh A, Desrivières S, Schumann G, Elliott P. New alcohol-related genes suggest shared genetic mechanisms with neuropsychiatric disorders. Nat Hum Behav 2019; 3:950-961. [PMID: 31358974 PMCID: PMC7711277 DOI: 10.1038/s41562-019-0653-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Excessive alcohol consumption is one of the main causes of death and disability worldwide. Alcohol consumption is a heritable complex trait. Here we conducted a meta-analysis of genome-wide association studies of alcohol consumption (g d-1) from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake. We identified 46 new common loci and investigated their potential functional importance using magnetic resonance imaging data and gene expression studies. We identify genetic pathways associated with alcohol consumption and suggest genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - He Gao
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Congying Chu
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Paul Blakeley
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, ITMAT Data Science Group, Imperial College London, London, UK
| | - Andrew R Butts
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Raha Pazoki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Hideaki Suzuki
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Andrianos M Yiorkas
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Ibrahim Karaman
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Joshua Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Department of Psychology and the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | | | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sebastian E Baumeister
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Chair of Epidemiology, Ludwig-Maximilians-Universitat Munchen, UNIKA-T Augsburg, Augsburg, Germany
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Toni-Kim Clarke
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Niki Dimou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A Kentistou
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas Foundation Trust, London, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mia-Maria Perälä
- Folkhälsan Research Center, Helsinki, Finland
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - David Porteous
- Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Scott M Ratliff
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Samuli Tuominen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alexis Wood
- Department of Pediatrics/Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juha Auvinen
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Oulunkaari Health Center, Ii, Finland
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Minna Männikkö
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Lorenz Risch
- Institute of Clinical Chemistry, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Labormedizinisches Zentrum Dr. Risch, Vaduz, Liechtenstein
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
- Department of Psychiatry, University Hospital of Oulu, Oulu, Finland
- Medical research Center Oulu, University and University Hospital of Oulu, Oulu, Finland
| | - Alexandra I Blakemore
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Catharina A Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Andrew C Heath
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Michael Kühne
- Cardiology Division, University Hospital Basel, Basel, Switzerland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
| | - Andrew M McIntosh
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Albertine J Oldehinkel
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | - Paul M Matthews
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Adrian Rothenfluh
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Departments of Psychiatry, Neurobiology & Anatomy, Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- PONS Research Group, Dept of Psychiatry and Psychotherapy, Campus Charite Mitte, Humboldt University, Berlin, Germany and Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, P.R. China.
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK.
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College Healthcare NHS Trust and Imperial College London, London, UK.
- Health Data Research UK London Substantive Site, London, UK.
| |
Collapse
|
24
|
Lim WJ, Kim KH, Kim JY, Kim HJ, Kim M, Park JL, Yoon S, Oh JH, Cho JW, Kim YS, Kim N. Investigation of Gene Expression and DNA Methylation From Seven Different Brain Regions of a Crab-Eating Monkey as Determined by RNA-Seq and Whole-Genome Bisulfite Sequencing. Front Genet 2019; 10:694. [PMID: 31428131 PMCID: PMC6690020 DOI: 10.3389/fgene.2019.00694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/02/2019] [Indexed: 01/04/2023] Open
Abstract
The crab-eating monkey is widely used in biomedical research for pharmacological experiments. Epigenetic regulation in the brain regions of primates involves complex patterns of DNA methylation. Previous studies of methylated CpG-binding domains using microarray technology or peak identification of sequence reads mostly focused on developmental stages or disease, rather than normal brains. To identify correlations between gene expression and DNA methylation levels that may be related to transcriptional regulation, we generated RNA-seq and whole-genome bisulfite sequencing data from seven different brain regions from a single crab-eating monkey. We identified 92 genes whose expression levels were significantly correlated, positively or negatively, with DNA methylation levels. Among them, 11 genes exhibited brain region-specific characteristics, and their expression patterns were strongly correlated with DNA methylation level. Nine genes (SLC2A5, MCM5, DRAM1, TTC12, DHX40, COR01A, LRAT, FLVCR2, and PTER) had effects on brain and eye function and development, and two (LHX6 and MEST) were previously identified as genes in which DNA methylation levels change significantly in the promoter region and are therefore considered brain epigenetic markers. Furthermore, we characterized DNA methylation of repetitive elements at the whole genome through repeat annotation at single-base resolution. Our results reveal the diverse roles of DNA methylation at single-base resolution throughout the genome and reflect the epigenetic variations in adult brain tissues.
Collapse
Affiliation(s)
- Won-Jun Lim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, South Korea
| | - Kyoung Hyoun Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, South Korea
| | - Jae-Yoon Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, South Korea
| | - Hee-Jin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Mirang Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jong-Lyul Park
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Seokjoo Yoon
- Predictive Toxicity Department, Korea Institute of Toxicology (KIT), Daejeon, South Korea
| | - Jung-Hwa Oh
- Predictive Toxicity Department, Korea Institute of Toxicology (KIT), Daejeon, South Korea
| | - Jae-Woo Cho
- Predictive Toxicity Department, Korea Institute of Toxicology (KIT), Daejeon, South Korea
| | - Yong Sung Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Namshin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
25
|
Macare C, Ducci F, Zhang Y, Ruggeri B, Jia T, Kaakinen M, Kalsi G, Charoen P, Casoni F, Peters J, Bromberg U, Hill M, Buxton J, Blakemore A, Veijola J, Büchel C, Banaschewski T, Bokde ALW, Conrod P, Flor H, Frouin V, Gallinat J, Garavan H, Gowland PA, Heinz A, Ittermann B, Lathrop M, Martinot JL, Paus T, Desrivières S, Munafò M, Järvelin MR, Schumann G. A neurobiological pathway to smoking in adolescence: TTC12-ANKK1-DRD2 variants and reward response. Eur Neuropsychopharmacol 2018; 28:1103-1114. [PMID: 30104163 PMCID: PMC6525784 DOI: 10.1016/j.euroneuro.2018.07.101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 06/13/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
The TTC12-ANKK1-DRD2 gene-cluster has been implicated in adult smoking. Here, we investigated the contribution of individual genes in the TTC12-ANKK1-DRD2 cluster in smoking and their association with smoking-associated reward processing in adolescence. A meta-analysis of TTC12-ANKK1-DRD2 variants and self-reported smoking behaviours was performed in four European adolescent cohorts (N = 14,084). The minor G-allele of rs2236709, mapping TTC12, was associated with self-reported smoking (p = 5.0 × 10-4) and higher plasma cotinine levels (p = 7.0 × 10-5). This risk allele was linked to an increased ventral-striatal blood-oxygen level-dependent (BOLD) response during reward anticipation (n = 1,263) and with higher DRD2 gene expression in the striatum (p = 0.013), but not with TTC12 or ANKK gene expression. These data suggest a role for the TTC12-ANKK1-DRD2 gene-cluster in adolescent smoking behaviours, provide evidence for the involvement of DRD2 in the early stages of addiction and support the notion that genetically-driven inter-individual differences in dopaminergic transmission mediate reward sensitivity and risk to smoking.
Collapse
Affiliation(s)
- Christine Macare
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Francesca Ducci
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; St George's, University of London, London, United Kingdom
| | - Yuning Zhang
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Barbara Ruggeri
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Tianye Jia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Marika Kaakinen
- Section of Genomics of Common Disease, Department of Medicine, Imperial College London, London, United Kingdom; Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, United Kingdom
| | - Gursharan Kalsi
- Clinical Physics, Barts Health NHS Trust, The Royal London Hospital, London, UK; One Small Step Gait Laboratory, Guy׳s and St. Thomas' NHS Foundation Trust, Guy׳s Hospital, London, UK
| | - Pimphen Charoen
- UCL Institute of Health Informatics, University College, London, UK
| | - Filippo Casoni
- Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy; Università Vita-Salute San Raffaele, Milan 20132, Italy
| | - Jan Peters
- Intelligent Autonomous Systems Group, Department of Computer Science, Technische Universität Darmstadt, Darmstadt, Germany; Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Uli Bromberg
- University Medical Centre Hamburg-Eppendorf, House W34, 3.OG, Martinistr. 52, 20246, Hamburg, Germany
| | - Matthew Hill
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jessica Buxton
- UCL Great Ormond Street Institute of Child Health, London; School of Life Sciences, Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| | - Alexandra Blakemore
- Division of Diabetes, Endocrinology and Metabolism, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland; Cambridge Cognition Ltd, Cambridge, UK
| | - Christian Büchel
- University Medical Centre Hamburg-Eppendorf, House W34, 3.OG, Martinistr. 52, 20246, Hamburg, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry, Central Institute of Mental Health, Faculty of Clinical Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Patricia Conrod
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Department of Psychiatry, Universite de Montreal, CHU Ste Justine Hospital, Montreal, Canada
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany; Department of Psychology, School of Social Sciences, University of Mannheim, 68131 Mannheim, Germany
| | | | - Jürgen Gallinat
- Clinic for Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405 Burlington, Vermont, USA
| | - Penny A Gowland
- Sir Peter Mansfield Magnetic Resonance Centre, University of Nottingham, Nottingham, United Kingdom
| | - Andreas Heinz
- Clinic for Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Mark Lathrop
- Department of Human Genetics, McGill University, 1205 Dr Penfield Avenue, Montreal, QC, Canada; McGill University, Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC, Canada
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM Unit 1000 "Neuroimaging & Psychiatry", University Paris Sud - Paris Saclay, University Paris Descartes; DIGITEO labs, Gif sur Yvette; France
| | - Tomáš Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital and Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Sylvane Desrivières
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Marcus Munafò
- MRC Integrative Epidemiology Unit, UK Centre for Tobacco Control Studies and School of Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - Marjo-Riitta Järvelin
- Institute of Health Sciences, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College London, United Kingdom; Unit of Primary Care, Oulu University Hospital, Oulu, Finland; Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland
| | - Gunter Schumann
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | | |
Collapse
|
26
|
Potenza MN. Clinical neuropsychiatric considerations regarding nonsubstance or behavioral addictions. DIALOGUES IN CLINICAL NEUROSCIENCE 2018. [PMID: 29302225 PMCID: PMC5741111 DOI: 10.31887/dcns.2017.19.3/mpotenza] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past several decades, non-substance-use behaviors like gambling, gaming, and sex have received greater consideration as possible foci of addictions. In this article, I will review the recent history and current status of non-substance or behavioral addictions. A main focus will involve gambling and gambling disorder, given that the latter is currently the sole non-substance addictive disorder described in the main text of the current (fifth) edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5). Internet gaming disorder, currently in the DSM-5 section addressing conditions that may need additional research, will also be considered, as will the concept of Internet addiction. Compulsive sexual behaviors (including problematic pornography use) will be considered, particularly with respect to how behavioral addictions may be considered in the forthcoming 11th edition of the International Classification of Diseases (ICD-11).
Collapse
Affiliation(s)
- Marc N Potenza
- Department of Psychiatry, Child Study Center, the National Center on Addiction and Substance Abuse, Yale University School of Medicine and the Connecticut Mental Health Center, Connecticut, USA
| |
Collapse
|
27
|
Pasman JA, Verweij KJH, Gerring Z, Stringer S, Sanchez-Roige S, Treur JL, Abdellaoui A, Nivard MG, Baselmans BML, Ong JS, Ip HF, van der Zee MD, Bartels M, Day FR, Fontanillas P, Elson SL, de Wit H, Davis LK, MacKillop J, Derringer JL, Branje SJT, Hartman CA, Heath AC, van Lier PAC, Madden PAF, Mägi R, Meeus W, Montgomery GW, Oldehinkel AJ, Pausova Z, Ramos-Quiroga JA, Paus T, Ribases M, Kaprio J, Boks MPM, Bell JT, Spector TD, Gelernter J, Boomsma DI, Martin NG, MacGregor S, Perry JRB, Palmer AA, Posthuma D, Munafò MR, Gillespie NA, Derks EM, Vink JM. GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal influence of schizophrenia. Nat Neurosci 2018; 21:1161-1170. [PMID: 30150663 PMCID: PMC6386176 DOI: 10.1038/s41593-018-0206-1] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/28/2018] [Indexed: 01/07/2023]
Abstract
Cannabis use is a heritable trait that has been associated with adverse mental health outcomes. In the largest genome-wide association study (GWAS) for lifetime cannabis use to date (N = 184,765), we identified eight genome-wide significant independent single nucleotide polymorphisms in six regions. All measured genetic variants combined explained 11% of the variance. Gene-based tests revealed 35 significant genes in 16 regions, and S-PrediXcan analyses showed that 21 genes had different expression levels for cannabis users versus nonusers. The strongest finding across the different analyses was CADM2, which has been associated with substance use and risk-taking. Significant genetic correlations were found with 14 of 25 tested substance use and mental health-related traits, including smoking, alcohol use, schizophrenia and risk-taking. Mendelian randomization analysis showed evidence for a causal positive influence of schizophrenia risk on cannabis use. Overall, our study provides new insights into the etiology of cannabis use and its relation with mental health.
Collapse
Affiliation(s)
- Joëlle A Pasman
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Karin J H Verweij
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, The Netherlands
| | - Zachary Gerring
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sven Stringer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Jorien L Treur
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Abdel Abdellaoui
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, The Netherlands
| | - Michel G Nivard
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bart M L Baselmans
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jue-Sheng Ong
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Hill F Ip
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Matthijs D van der Zee
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Meike Bartels
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Felix R Day
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Lea K Davis
- Vanderbilt Genetics Institute; Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - James MacKillop
- Peter Boris Centre for Addictions Research and Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University/St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Jaime L Derringer
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Susan J T Branje
- Department of Youth and Family, Utrecht University, Utrecht, the Netherlands
| | - Catharina A Hartman
- Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Pol A C van Lier
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pamela A F Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Wim Meeus
- Department of Youth and Family, Utrecht University, Utrecht, the Netherlands
| | - Grant W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - A J Oldehinkel
- Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Josep A Ramos-Quiroga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tomas Paus
- Rotman Research Institute, Baycrest, Toronto, Ontario, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Marta Ribases
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, HiLIFE Unit, University of Helsinki, Helsinki, Finland
| | - Marco P M Boks
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Dorret I Boomsma
- Department of Biological Psychology/Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nicholas G Martin
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Stuart MacGregor
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - 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
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies and School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Nathan A Gillespie
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Eske M Derks
- Genetic Epidemiology, Statistical Genetics, and Translational Neurogenomics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jacqueline M Vink
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Al-Obaide MAI, Ibrahim BA, Al-Humaish S, Abdel-Salam ASG. Genomic and Bioinformatics Approaches for Analysis of Genes Associated With Cancer Risks Following Exposure to Tobacco Smoking. Front Public Health 2018; 6:84. [PMID: 29616208 PMCID: PMC5869936 DOI: 10.3389/fpubh.2018.00084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/05/2018] [Indexed: 01/03/2023] Open
Abstract
Cancer is a significant health problem in the Middle East and global population. It is well established that there is a direct link between tobacco smoking and cancer, which will continue to pose a significant threat to human health. The impact of long-term exposure to tobacco smoke on the risk of cancer encouraged the study of biomarkers for vulnerable individuals to tobacco smoking, especially children, who are more susceptible than adults to the action of environmental carcinogens. The carcinogens in tobacco smoke condensate induce DNA damage and play a significant role in determining the health and well-being of smokers, non-smoker, and primarily children. Cancer is a result of genomic and epigenomic malfunctions that lead to an initial premalignant condition. Although premalignancy genetic cascade is a much-delayed process, it will end with adverse health consequences. In addition to the DNA damage and mutations, tobacco smoke can cause changes in the DNA methylation and gene expression associated with cancer. The genetic events hint on the possible use of genomic–epigenomic changes in genes related to cancer, in predicting cancer risks associated with exposure to tobacco smoking. Bioinformatics provides indispensable tools to identify the cascade of expressed genes in active smokers and non-smokers and could assist the development of a framework to manage this cascade of events linked with the evolvement of disease including cancer. The aim of this mini review is to cognize the essential genomic processes and health risks associated with tobacco smoking and the implications of bioinformatics in cancer prediction, prevention, and intervention.
Collapse
Affiliation(s)
- Mohammed A I Al-Obaide
- Department of Biomedical Science, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, TX, United States
| | | | | | - Abdel-Salam G Abdel-Salam
- Department of Mathematics, Statistics and Physics, College of Arts and Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
29
|
The Role of Cell Adhesion Molecule Genes Regulating Neuroplasticity in Addiction. Neural Plast 2018; 2018:9803764. [PMID: 29675039 PMCID: PMC5838467 DOI: 10.1155/2018/9803764] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/10/2017] [Indexed: 01/06/2023] Open
Abstract
A variety of genetic approaches, including twin studies, linkage studies, and candidate gene studies, has established a firm genetic basis for addiction. However, there has been difficulty identifying the precise genes that underlie addiction liability using these approaches. This situation became especially clear in genome-wide association studies (GWAS) of addiction. Moreover, the results of GWAS brought into clarity many of the shortcomings of those early genetic approaches. GWAS studies stripped away those preconceived notions, examining genes that would not previously have been considered in the study of addiction, consequently creating a shift in our understanding. Most importantly, those studies implicated a class of genes that had not previously been considered in the study of addiction genetics: cell adhesion molecules (CAMs). Considering the well-documented evidence supporting a role for various CAMs in synaptic plasticity, axonal growth, and regeneration, it is not surprising that allelic variation in CAM genes might also play a role in addiction liability. This review focuses on the role of various cell adhesion molecules in neuroplasticity that might contribute to addictive processes and emphasizes the importance of ongoing research on CAM genes that have been implicated in addiction by GWAS.
Collapse
|
30
|
Prom-Wormley EC, Ebejer J, Dick DM, Bowers MS. The genetic epidemiology of substance use disorder: A review. Drug Alcohol Depend 2017; 180:241-259. [PMID: 28938182 PMCID: PMC5911369 DOI: 10.1016/j.drugalcdep.2017.06.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Substance use disorder (SUD) remains a significant public health issue. A greater understanding of how genes and environment interact to regulate phenotypes comprising SUD will facilitate directed treatments and prevention. METHODS The literature studying the neurobiological correlates of SUD with a focus on the genetic and environmental influences underlying these mechanisms was reviewed. Results from twin/family, human genetic association, gene-environment interaction, epigenetic literature, phenome-wide association studies are summarized for alcohol, nicotine, cannabinoids, cocaine, and opioids. RESULTS There are substantial genetic influences on SUD that are expected to influence multiple neurotransmission pathways, and these influences are particularly important within the dopaminergic system. Genetic influences involved in other aspects of SUD etiology including drug processing and metabolism are also identified. Studies of gene-environment interaction emphasize the importance of environmental context in SUD. Epigenetic studies indicate drug-specific changes in gene expression as well as differences in gene expression related to the use of multiple substances. Further, gene expression is expected to differ by stage of SUD such as substance initiation versus chronic substance use. While a substantial literature has developed for alcohol and nicotine use disorders, there is comparatively less information for other commonly abused substances. CONCLUSIONS A better understanding of genetically-mediated mechanisms involved in the neurobiology of SUD provides increased opportunity to develop behavioral and biologically based treatment and prevention of SUD.
Collapse
Affiliation(s)
- Elizabeth C Prom-Wormley
- Dvision of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, PO Box 980212, Richmond, VA 23298-0212, USA.
| | - Jane Ebejer
- School of Cognitive Behavioural and Social Sciences, University of New England, Armidale, NSW 2350, Australia
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, PO Box 842509, Richmond, VA 23284-2509, USA
| | - M Scott Bowers
- Faulk Center for Molecular Therapeutics, Biomedical Engeneering, Northwestern University, Evanston, IL 60201, USA
| |
Collapse
|
31
|
Further replication of the synergistic interaction between LPHN3 and the NTAD gene cluster on ADHD and its clinical course throughout adulthood. Prog Neuropsychopharmacol Biol Psychiatry 2017. [PMID: 28624582 DOI: 10.1016/j.pnpbp.2017.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is a common and highly heritable neuropsychiatric disorder. Despite the high heritability, the unraveling of specific genetic factors related to ADHD is hampered by its considerable genetic complexity. Recent evidence suggests that gene-gene interactions can explain part of this complexity. We examined the impact of strongly supported interaction effects between the LPHN3 gene and the NTAD gene cluster (NCAM1-TTC12-ANKK1-DRD2) in a 7-year follow-up of a clinical sample of adults with ADHD, addressing associations with susceptibility, symptomatology and stability of diagnosis. The sample comprises 548 adults with ADHD and 643 controls. Entropy-based analysis indicated a potential interaction between the LPHN3-rs6551665 and TTC12-rs2303380 SNPs influencing ADHD symptom counts. Further analyses revealed significant interaction effects on ADHD total symptoms (p=0.002), and with hyperactivity/impulsivity symptom counts (p=0.005). In the group composed by predominantly hyperactive/impulsive and combined presentation, the presence of LPHN3-rs6551665 G allele was related to increased ADHD risk only in individuals carrying the TTC12-rs2303380 AA genotype (p=0.026). Also, the same allelic constellation is involved in maintenance of ADHD in a predominantly hyperactive/impulsive or combined presentation after a 7-year follow-up (p=0.008). These observations reinforce and replicate previous evidence suggesting that an interaction effect between the LPHN3 gene and the NTAD cluster may have a role in the genetic substrate associated to ADHD also in adults. Moreover, it is possible that the interactions between LPHN3 and NTAD are specific factors contributing to the development of an ADHD phenotype with increased hyperactivity/impulsivity that is maintained throughout adulthood.
Collapse
|
32
|
Lippard ETC, Jensen KP, Wang F, Johnston JAY, Spencer L, Pittman B, Gelernter J, Blumberg HP. Effects of ANK3 variation on gray and white matter in bipolar disorder. Mol Psychiatry 2017; 22:1345-1351. [PMID: 27240527 PMCID: PMC5133179 DOI: 10.1038/mp.2016.76] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/23/2016] [Accepted: 04/05/2016] [Indexed: 01/22/2023]
Abstract
The single-nucleotide polymorphism rs9804190 in the Ankyrin G (ANK3) gene has been reported in genome-wide association studies to be associated with bipolar disorder (BD). However, the neural system effects of rs9804190 in BD are not known. We investigated associations between rs9804190 and gray and white matter (GM and WM, respectively) structure within a frontotemporal neural system implicated in BD. A total of 187 adolescent and adult European Americans were studied: a group homozygous for the C allele (52 individuals with BD and 56 controls) and a T-carrier group, carrying the high-risk T allele (38 BD and 41 controls). Subjects participated in high-resolution structural magnetic resonance imaging and diffusion tensor imaging (DTI) scanning. Frontotemporal region of interest (ROI) and whole-brain exploratory analyses were conducted. DTI ROI-based analysis revealed a significant diagnosis by genotype interaction within the uncinate fasciculus (P⩽0.05), with BD subjects carrying the T (risk) allele showing decreased fractional anisotropy compared with other subgroups, independent of age. Genotype effects were not observed in frontotemporal GM volume. These findings support effects of rs9804190 on frontotemporal WM in adolescents and adults with BD and suggest a mechanism contributing to WM pathology in BD.
Collapse
Affiliation(s)
- E T C Lippard
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - K P Jensen
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, VA Connecticut Healthcare System, West Haven, CT, USA
| | - F Wang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - J A Y Johnston
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - L Spencer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - B Pittman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - J Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, VA Connecticut Healthcare System, West Haven, CT, USA
| | - H P Blumberg
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
33
|
Richter A, Barman A, Wüstenberg T, Soch J, Schanze D, Deibele A, Behnisch G, Assmann A, Klein M, Zenker M, Seidenbecher C, Schott BH. Behavioral and Neural Manifestations of Reward Memory in Carriers of Low-Expressing versus High-Expressing Genetic Variants of the Dopamine D2 Receptor. Front Psychol 2017; 8:654. [PMID: 28507526 PMCID: PMC5410587 DOI: 10.3389/fpsyg.2017.00654] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/12/2017] [Indexed: 12/13/2022] Open
Abstract
Dopamine is critically important in the neural manifestation of motivated behavior, and alterations in the human dopaminergic system have been implicated in the etiology of motivation-related psychiatric disorders, most prominently addiction. Patients with chronic addiction exhibit reduced dopamine D2 receptor (DRD2) availability in the striatum, and the DRD2 TaqIA (rs1800497) and C957T (rs6277) genetic polymorphisms have previously been linked to individual differences in striatal dopamine metabolism and clinical risk for alcohol and nicotine dependence. Here, we investigated the hypothesis that the variants of these polymorphisms would show increased reward-related memory formation, which has previously been shown to jointly engage the mesolimbic dopaminergic system and the hippocampus, as a potential intermediate phenotype for addiction memory. To this end, we performed functional magnetic resonance imaging (fMRI) in 62 young, healthy individuals genotyped for DRD2 TaqIA and C957T variants. Participants performed an incentive delay task, followed by a recognition memory task 24 h later. We observed effects of both genotypes on the overall recognition performance with carriers of low-expressing variants, namely TaqIA A1 carriers and C957T C homozygotes, showing better performance than the other genotype groups. In addition to the better memory performance, C957T C homozygotes also exhibited a response bias for cues predicting monetary reward. At the neural level, the C957T polymorphism was associated with a genotype-related modulation of right hippocampal and striatal fMRI responses predictive of subsequent recognition confidence for reward-predicting items. Our results indicate that genetic variations associated with DRD2 expression affect explicit memory, specifically for rewarded stimuli. We suggest that the relatively better memory for rewarded stimuli in carriers of low-expressing DRD2 variants may reflect an intermediate phenotype of addiction memory.
Collapse
Affiliation(s)
- Anni Richter
- Leibniz Institute for NeurobiologyMagdeburg, Germany
| | | | - Torsten Wüstenberg
- Department of Psychiatry and Psychotherapy, Charité University HospitalBerlin, Germany
| | - Joram Soch
- Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Denny Schanze
- Institute of Human Genetics, Otto von Guericke UniversityMagdeburg, Germany
| | - Anna Deibele
- Leibniz Institute for NeurobiologyMagdeburg, Germany
| | | | - Anne Assmann
- Leibniz Institute for NeurobiologyMagdeburg, Germany.,Department of Neurology, University of MagdeburgMagdeburg, Germany
| | - Marieke Klein
- Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, Otto von Guericke UniversityMagdeburg, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for NeurobiologyMagdeburg, Germany.,Center for Behavioral Brain SciencesMagdeburg, Germany
| | - Björn H Schott
- Leibniz Institute for NeurobiologyMagdeburg, Germany.,Department of Psychiatry and Psychotherapy, Charité University HospitalBerlin, Germany.,Department of Neurology, University of MagdeburgMagdeburg, Germany.,Center for Behavioral Brain SciencesMagdeburg, Germany
| |
Collapse
|
34
|
Smith CT, Dang LC, Buckholtz JW, Tetreault AM, Cowan RL, Kessler RM, Zald DH. The impact of common dopamine D2 receptor gene polymorphisms on D2/3 receptor availability: C957T as a key determinant in putamen and ventral striatum. Transl Psychiatry 2017; 7:e1091. [PMID: 28398340 PMCID: PMC5416688 DOI: 10.1038/tp.2017.45] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/02/2016] [Accepted: 01/17/2017] [Indexed: 12/20/2022] Open
Abstract
Dopamine function is broadly implicated in multiple neuropsychiatric conditions believed to have a genetic basis. Although a few positron emission tomography (PET) studies have investigated the impact of single-nucleotide polymorphisms (SNPs) in the dopamine D2 receptor gene (DRD2) on D2/3 receptor availability (binding potential, BPND), these studies have often been limited by small sample size. Furthermore, the most commonly studied SNP in D2/3 BPND (Taq1A) is not located in the DRD2 gene itself, suggesting that its linkage with other DRD2 SNPs may explain previous PET findings. Here, in the largest PET genetic study to date (n=84), we tested for effects of the C957T and -141C Ins/Del SNPs (located within DRD2) as well as Taq1A on BPND of the high-affinity D2 receptor tracer 18F-Fallypride. In a whole-brain voxelwise analysis, we found a positive linear effect of C957T T allele status on striatal BPND bilaterally. The multilocus genetic scores containing C957T and one or both of the other SNPs produced qualitatively similar striatal results to C957T alone. The number of C957T T alleles predicted BPND in anatomically defined putamen and ventral striatum (but not caudate) regions of interest, suggesting some regional specificity of effects in the striatum. By contrast, no significant effects arose in cortical regions. Taken together, our data support the critical role of C957T in striatal D2/3 receptor availability. This work has implications for a number of psychiatric conditions in which dopamine signaling and variation in C957T status have been implicated, including schizophrenia and substance use disorders.
Collapse
Affiliation(s)
- C T Smith
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - L C Dang
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - J W Buckholtz
- Department of Psychology, Harvard University, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - A M Tetreault
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - R L Cowan
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - R M Kessler
- Department of Radiology, UAB School of Medicine, Birmingham, AL, USA
| | - D H Zald
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
35
|
Hodgson K, Almasy L, Knowles EEM, Kent JW, Curran JE, Dyer TD, Göring HHH, Olvera RL, Woolsey MD, Duggirala R, Fox PT, Blangero J, Glahn DC. The genetic basis of the comorbidity between cannabis use and major depression. Addiction 2017; 112:113-123. [PMID: 27517884 PMCID: PMC5148647 DOI: 10.1111/add.13558] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/06/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS While the prevalence of major depression is elevated among cannabis users, the role of genetics in this pattern of comorbidity is not clear. This study aimed to estimate the heritability of cannabis use and major depression, quantify the genetic overlap between these two traits and localize regions of the genome that segregate in families with cannabis use and major depression. DESIGN Family-based univariate and bivariate genetic analysis. SETTING San Antonio, Texas, USA. PARTICIPANTS Genetics of Brain Structure and Function study (GOBS) participants: 1284 Mexican Americans from 75 large multi-generation families and an additional 57 genetically unrelated spouses. MEASUREMENTS Phenotypes of life-time history of cannabis use and major depression, measured using the semistructured MINI-Plus interview. Genotypes measured using ~1 M single nucleotide polymorphisms (SNPs) on Illumina BeadChips. A subselection of these SNPs were used to build multi-point identity-by-descent matrices for linkage analysis. FINDINGS Both cannabis use [h2 = 0.614, P = 1.00 × 10-6 , standard error (SE) = 0.151] and major depression (h2 = 0.349, P = 1.06 × 10-5 , SE = 0.100) are heritable traits, and there is significant genetic correlation between the two (ρg = 0.424, P = 0.0364, SE = 0.195). Genome-wide linkage scans identify a significant univariate linkage peak for major depression on chromosome 22 [logarithm of the odds (LOD) = 3.144 at 2 centimorgans (cM)], with a suggestive peak for cannabis use on chromosome 21 (LOD = 2.123 at 37 cM). A significant pleiotropic linkage peak influencing both cannabis use and major depression was identified on chromosome 11 using a bivariate model (LOD = 3.229 at 112 cM). Follow-up of this pleiotropic signal identified a SNP 20 kb upstream of NCAM1 (rs7932341) that shows significant bivariate association (P = 3.10 × 10-5 ). However, this SNP is rare (seven minor allele carriers) and does not drive the linkage signal observed. CONCLUSIONS There appears to be a significant genetic overlap between cannabis use and major depression among Mexican Americans, a pleiotropy that appears to be localized to a region on chromosome 11q23 that has been linked previously to these phenotypes.
Collapse
Affiliation(s)
- Karen Hodgson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT,Correspondence to Karen Hodgson Ph.D., Department of Psychiatry, Yale School of Medicine, New Haven, CT
| | - Laura Almasy
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - Emma E. M. Knowles
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Jack W. Kent
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - Thomas D. Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - Harald H. H. Göring
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - Rene L. Olvera
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Mary D. Woolsey
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Ravi Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - Peter T. Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, South Texas Veterans Health System, 7400 Merton Minter, San Antonio, TX
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Health Science Center at San Antonio & University of Texas of the Rio Grande Valley, Brownsville, TX
| | - David C. Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT
| |
Collapse
|
36
|
Converging findings from linkage and association analyses on susceptibility genes for smoking and other addictions. Mol Psychiatry 2016; 21:992-1008. [PMID: 27166759 PMCID: PMC4956568 DOI: 10.1038/mp.2016.67] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 12/18/2022]
Abstract
Experimental approaches to genetic studies of complex traits evolve with technological advances. How do discoveries using different approaches advance our knowledge of the genetic architecture underlying complex diseases/traits? Do most of the findings of newer techniques, such as genome-wide association study (GWAS), provide more information than older ones, for example, genome-wide linkage study? In this review, we address these issues by developing a nicotine dependence (ND) genetic susceptibility map based on the results obtained by the approaches commonly used in recent years, namely, genome-wide linkage, candidate gene association, GWAS and targeted sequencing. Converging and diverging results from these empirical approaches have elucidated a preliminary genetic architecture of this intractable psychiatric disorder and yielded new hypotheses on ND etiology. The insights we obtained by putting together results from diverse approaches can be applied to other complex diseases/traits. In sum, developing a genetic susceptibility map and keeping it updated are effective ways to keep track of what we know about a disease/trait and what the next steps may be with new approaches.
Collapse
|
37
|
Nagy V, Cole T, Van Campenhout C, Khoung TM, Leung C, Vermeiren S, Novatchkova M, Wenzel D, Cikes D, Polyansky AA, Kozieradzki I, Meixner A, Bellefroid EJ, Neely GG, Penninger JM. The evolutionarily conserved transcription factor PRDM12 controls sensory neuron development and pain perception. Cell Cycle 2016; 14:1799-808. [PMID: 25891934 DOI: 10.1080/15384101.2015.1036209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PR homology domain-containing member 12 (PRDM12) belongs to a family of conserved transcription factors implicated in cell fate decisions. Here we show that PRDM12 is a key regulator of sensory neuronal specification in Xenopus. Modeling of human PRDM12 mutations that cause hereditary sensory and autonomic neuropathy (HSAN) revealed remarkable conservation of the mutated residues in evolution. Expression of wild-type human PRDM12 in Xenopus induced the expression of sensory neuronal markers, which was reduced using various human PRDM12 mutants. In Drosophila, we identified Hamlet as the functional PRDM12 homolog that controls nociceptive behavior in sensory neurons. Furthermore, expression analysis of human patient fibroblasts with PRDM12 mutations uncovered possible downstream target genes. Knockdown of several of these target genes including thyrotropin-releasing hormone degrading enzyme (TRHDE) in Drosophila sensory neurons resulted in altered cellular morphology and impaired nociception. These data show that PRDM12 and its functional fly homolog Hamlet are evolutionary conserved master regulators of sensory neuronal specification and play a critical role in pain perception. Our data also uncover novel pathways in multiple species that regulate evolutionary conserved nociception.
Collapse
Key Words
- BSA, bovine serum albumin
- Brn3d, brain 3d
- CGNL1, cyclin L1
- ChIP, chromatin immunoprecipitation
- DAPI, 4′,6-diamidino-2-phenylindole
- DDK, DYKDDDDK epitope
- Drgx, dorsal root ganglia homeobox
- ECL, enhanced chemiluminescence
- En1, engrailed-1
- FDR, false discovery rate
- FPKM, fragments per kilobase exon
- GAPDH, glyceraldehyde 3-phospate dehydrogenase
- GEO, gene expression omnibus
- GFP, green fluorescent protein
- HEK293, human embryonic kidney cell 293
- HRP, horseraddish peroxidase
- HSAN, hereditary and sensory autonomic neuropathy
- Hamlet
- Hmx3, H6 family homeobox 3
- IL1R1, interleukin 1 receptor type 1
- MO, morpholino oligonucleotide
- NBT/BCIP, nitro blue tetrazolium / 5-bromo-4-chloro-3-indolyl-phosphate
- PBS, phosphate buffered saline
- PDB, protein data base
- PMID, pubmed identification.
- PRDM12
- PRDM12, PR homology domain-containing member 12
- RA, retinoic acid
- RT-qPCR, real-time quantitative polymerase chain reaction
- S1PR1, Sphi8ngosine-1-phosphate receptor 1
- SET, Su(var)3–9 and ‘Enhancer of zeste’
- Sncg, Synuclein Gamma (Breast Cancer-Specific Protein 1)
- TRH(DE), tryrotropin-releasing hormone degrading enzyme
- TRHDE
- TRHDE, tyrotropin-releasing hormone degrading enzyme
- Tlx3, T-cell leukemia homeobox 3
- nociception
- pCMV6, plasmid cytomegalovirus
- sensory neurons
Collapse
Affiliation(s)
- Vanja Nagy
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria; UNSW Medicine, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cartmel B, Dewan A, Ferrucci LM, Gelernter J, Stapleton J, Leffell DJ, Mayne ST, Bale AE. Novel gene identified in an exome-wide association study of tanning dependence. Exp Dermatol 2016; 23:757-9. [PMID: 25041255 DOI: 10.1111/exd.12503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2014] [Indexed: 01/01/2023]
Abstract
Growing evidence suggests that some individuals may exhibit symptoms of dependence to ultraviolet light, a known carcinogen, in the context of tanning. Genetic associations with tanning dependence (TD) have not yet been explored. We conducted an exome-wide association study in 79 individuals who exhibited symptoms of TD and 213 individuals with volitional exposure to ultraviolet light, but who were not TD based on three TD scales. A total of 300 000 mostly exomic single nucleotide polymorphisms primarily in coding regions were assessed using an Affymetrix Axiom array. We performed a gene burden test with Bonferroni correction for the number of genes examined (P < 0.05/14 904 = 3.36 × 10(-6) ). One gene, patched domain containing 2 (PTCHD2), yielded a statistically significant P-value of 2.5 × 10(-6) (OR = 0.27) with fewer individuals classified as TD having a minor allele at this locus. These results require replication, but are the first to support a specific genetic association with TD.
Collapse
Affiliation(s)
- Brenda Cartmel
- Yale School of Public Health, New Haven, CT, USA; Yale Cancer Center, New Haven, CT, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hill SY, Jones BL, Steinhauer SR, Zezza N, Stiffler S. Longitudinal predictors of cannabis use and dependence in offspring from families at ultra high risk for alcohol dependence and in control families. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:383-95. [PMID: 26756393 PMCID: PMC5444658 DOI: 10.1002/ajmg.b.32417] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 01/04/2016] [Indexed: 11/06/2022]
Abstract
Cannabis use is common among adolescents. Identification of the factors associated with continued heavy use into young adulthood and development of cannabis abuse and dependence is of considerable importance. The role of familial risk for addiction and an associated endophenotype, P300 amplitude, has not previously been related to cannabis use and dependence. A prospective longitudinal study spanning childhood and young adulthood provided the opportunity for exploring these factors, along with genetic variation, in the cannabis use behaviors of 338 young adult offspring from high and low familial risk for alcohol dependence families (ages 19-30). P300 data were collected multiple times in childhood. The association between young adult patterns of cannabis use or cannabis abuse/dependence was tested with genetic variation in the cannabinoid gene, CNR1, the ANKK1-DRD2 gene, and childhood developmental trajectories of P300. Young adult patterns of cannabis use was characterized by three patterns: (i) no use throughout; (ii) declining use from adolescence through young adulthood; and (iii) frequent use throughout. Following the low P300 trajectory in childhood predicted cannabis abuse and dependence by young adulthood. A four SNP ANKK1-DRD2 haplotype (G-G-G-C) was found to be significantly associated with the frequency of use patterns (P = 0.0008). Although CNR1 variation overall was not significantly associated with these patterns, among individuals with cannabis abuse/dependence the presence of one or both copies of the rs806368 A > G minor allele conferred a 5.4-fold increase (P = 0.003) in the likelihood that they would be in the frequent and persistent use group rather than the declining use group.
Collapse
Affiliation(s)
- Shirley Y. Hill
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Correspondence to: Shirley Y. Hill, Ph.D., Department of Psychiatry, University of Pittsburgh Medical Center, 3811 O’ Hara St. Pittsburgh, PA 15213.
| | - Bobby L. Jones
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Nicholas Zezza
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Scott Stiffler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
40
|
Esterlis I, Hillmer AT, Bois F, Pittman B, McGovern E, O'Malley SS, Picciotto MR, Yang BZ, Gelernter J, Cosgrove KP. CHRNA4 and ANKK1 Polymorphisms Influence Smoking-Induced Nicotinic Acetylcholine Receptor Upregulation. Nicotine Tob Res 2016; 18:1845-52. [PMID: 27611310 DOI: 10.1093/ntr/ntw081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/22/2016] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Tobacco smoking leads to increased numbers of β2*-containing nicotinic acetylcholine receptors (β2*-nAChRs) throughout the brain, which return to nonsmoker levels over extended abstinence. The goal of the current study was to determine whether the degree of tobacco smoking-induced changes in β2*-nAChR availability is genetically influenced. METHODS In this study, 113 European Americans participated in one or two [(123)I]5-IA-85380 single photon emission computed tomography (SPECT) brain scans. Smokers (n = 58) participated in one scan at 7-9 days of abstinence and those who remained abstinent (n = 27) were imaged again at 6-8 weeks of abstinence. Age- and sex-matched nonsmokers (n = 55) participated in one scan. Blood samples were collected for DNA analysis and genotyped for single nucleotide polymorphisms (SNPs) in the CHRNA4 and ANKK1 gene loci. β2*-nAChR availability was measured in the thalamus, striatum, cortical regions, and cerebellum. RESULTS The CHRNA4 SNP rs2236196 and ANKK1 SNP rs4938015 were associated with significantly higher cerebellar and cortical β2*-nAChR availability in smokers versus nonsmokers for specific genotypes. There were no significant differences by carrier status in the change in β2*-nAChR availability in smokers from 7-9 days to 6-8 weeks of abstinence. CONCLUSION This study provides evidence for genetic regulation of tobacco smoking-induced changes in β2*-nAChR availability and suggests that β2*-nAChR availability could be an endophenotype mediating influences of CHRNA4 variants on nicotine dependence. These results highlight individual differences in the neurochemistry of nicotine dependence and may suggest the need for individualized programs for smoking cessation. IMPLICATIONS This study demonstrates genetic regulation of smoking-induced changes in β2*-nAChRs throughout the brain and highlights the need for personalized programs for smoking cessation.
Collapse
Affiliation(s)
- Irina Esterlis
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT; Department of Radiology, Yale University School of Medicine, New Haven, CT
| | - Ansel T Hillmer
- Department of Radiology, Yale University School of Medicine, New Haven, CT
| | - Frederic Bois
- Department of Radiology, Yale University School of Medicine, New Haven, CT
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Erin McGovern
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | | | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT; Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT; Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT; Department of Radiology, Yale University School of Medicine, New Haven, CT;
| |
Collapse
|
41
|
Stringer S, Minică CC, Verweij KJH, Mbarek H, Bernard M, Derringer J, van Eijk KR, Isen JD, Loukola A, Maciejewski DF, Mihailov E, van der Most PJ, Sánchez-Mora C, Roos L, Sherva R, Walters R, Ware JJ, Abdellaoui A, Bigdeli TB, Branje SJT, Brown SA, Bruinenberg M, Casas M, Esko T, Garcia-Martinez I, Gordon SD, Harris JM, Hartman CA, Henders AK, Heath AC, Hickie IB, Hickman M, Hopfer CJ, Hottenga JJ, Huizink AC, Irons DE, Kahn RS, Korhonen T, Kranzler HR, Krauter K, van Lier PAC, Lubke GH, Madden PAF, Mägi R, McGue MK, Medland SE, Meeus WHJ, Miller MB, Montgomery GW, Nivard MG, Nolte IM, Oldehinkel AJ, Pausova Z, Qaiser B, Quaye L, Ramos-Quiroga JA, Richarte V, Rose RJ, Shin J, Stallings MC, Stiby AI, Wall TL, Wright MJ, Koot HM, Paus T, Hewitt JK, Ribasés M, Kaprio J, Boks MP, Snieder H, Spector T, Munafò MR, Metspalu A, Gelernter J, Boomsma DI, Iacono WG, Martin NG, Gillespie NA, Derks EM, Vink JM. Genome-wide association study of lifetime cannabis use based on a large meta-analytic sample of 32 330 subjects from the International Cannabis Consortium. Transl Psychiatry 2016; 6:e769. [PMID: 27023175 PMCID: PMC4872459 DOI: 10.1038/tp.2016.36] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 01/15/2023] Open
Abstract
Cannabis is the most widely produced and consumed illicit psychoactive substance worldwide. Occasional cannabis use can progress to frequent use, abuse and dependence with all known adverse physical, psychological and social consequences. Individual differences in cannabis initiation are heritable (40-48%). The International Cannabis Consortium was established with the aim to identify genetic risk variants of cannabis use. We conducted a meta-analysis of genome-wide association data of 13 cohorts (N=32 330) and four replication samples (N=5627). In addition, we performed a gene-based test of association, estimated single-nucleotide polymorphism (SNP)-based heritability and explored the genetic correlation between lifetime cannabis use and cigarette use using LD score regression. No individual SNPs reached genome-wide significance. Nonetheless, gene-based tests identified four genes significantly associated with lifetime cannabis use: NCAM1, CADM2, SCOC and KCNT2. Previous studies reported associations of NCAM1 with cigarette smoking and other substance use, and those of CADM2 with body mass index, processing speed and autism disorders, which are phenotypes previously reported to be associated with cannabis use. Furthermore, we showed that, combined across the genome, all common SNPs explained 13-20% (P<0.001) of the liability of lifetime cannabis use. Finally, there was a strong genetic correlation (rg=0.83; P=1.85 × 10(-8)) between lifetime cannabis use and lifetime cigarette smoking implying that the SNP effect sizes of the two traits are highly correlated. This is the largest meta-analysis of cannabis GWA studies to date, revealing important new insights into the genetic pathways of lifetime cannabis use. Future functional studies should explore the impact of the identified genes on the biological mechanisms of cannabis use.
Collapse
Affiliation(s)
- S Stringer
- Department of Complex Trait Genetics, VU Amsterdam, Center for Neurogenomics and Cognitive Research, Amsterdam, The Netherlands
- Department of Psychiatry, Academic Medical Centre, Amsterdam, The Netherlands
| | - C C Minică
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - K J H Verweij
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, VU University, Amsterdam, The Netherlands
| | - H Mbarek
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - M Bernard
- The Hospital for Sick Children Research Institute, Toronto, Canada
| | - J Derringer
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - K R van Eijk
- Department of Human Neurogenetics, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J D Isen
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - A Loukola
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
| | - D F Maciejewski
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, VU University, Amsterdam, The Netherlands
| | - E Mihailov
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - P J van der Most
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - C Sánchez-Mora
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Spain
| | - L Roos
- Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - R Sherva
- Biomedical Genetics Department, Boston University School of Medicine, Boston, MA, USA
| | - R Walters
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - J J Ware
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - A Abdellaoui
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - T B Bigdeli
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - S J T Branje
- Research Centre Adolescent Development, Utrecht University, Utrecht, The Netherlands
| | - S A Brown
- Department of Psychology and Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - M Bruinenberg
- The LifeLines Cohort Study, University of Groningen, Groningen, The Netherlands
| | - M Casas
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - T Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - I Garcia-Martinez
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - S D Gordon
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - J M Harris
- Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - C A Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A K Henders
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - A C Heath
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - I B Hickie
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - M Hickman
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - C J Hopfer
- Department of Psychiatry, University of Colorado Denver, Aurora, CO, USA
| | - J J Hottenga
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - A C Huizink
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, VU University, Amsterdam, The Netherlands
| | - D E Irons
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - R S Kahn
- Department of Human Neurogenetics, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - T Korhonen
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | - H R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - K Krauter
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - P A C van Lier
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, VU University, Amsterdam, The Netherlands
| | - G H Lubke
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
- Department of Psychology, University of Notre Dame, Notre Dame, IN, USA
| | - P A F Madden
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - R Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - M K McGue
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - S E Medland
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - W H J Meeus
- Research Centre Adolescent Development, Utrecht University, Utrecht, The Netherlands
- Developmental Psychology, Tilburg University, Tilburg, The Netherlands
| | - M B Miller
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - G W Montgomery
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - M G Nivard
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - I M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A J Oldehinkel
- Interdisciplinary Center for Pathology and Emotion Regulation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Z Pausova
- The Hospital for Sick Children Research Institute, Toronto, Canada
- Department of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - B Qaiser
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
| | - L Quaye
- Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - J A Ramos-Quiroga
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - V Richarte
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - R J Rose
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, USA
| | - J Shin
- The Hospital for Sick Children Research Institute, Toronto, Canada
| | - M C Stallings
- Department of Psychology and Neuroscience, Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - A I Stiby
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - T L Wall
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - M J Wright
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - H M Koot
- Department of Developmental Psychology and EMGO Institute for Health and Care Research, VU University, Amsterdam, The Netherlands
| | - T Paus
- Rotman Research Institute, Baycrest, Toronto, ON, Canada
- Department of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA
| | - J K Hewitt
- Department of Psychology and Neuroscience, Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - M Ribasés
- Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Barcelona, Spain
| | - J Kaprio
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - M P Boks
- Department of Human Neurogenetics, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - T Spector
- Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - M R Munafò
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies and School of Experimental Psychology, University of Bristol, Bristol, UK
| | - A Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - J Gelernter
- Department of Psychiatry, Genetics, and Neurobiology, Yale University School of Medicine and VA CT, West Haven, CT, USA
| | - D I Boomsma
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
- Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - W G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - N G Martin
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - N A Gillespie
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Genetic Epidemiology, Molecular Epidemiology and Neurogenetics Laboratories, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - E M Derks
- Department of Psychiatry, Academic Medical Centre, Amsterdam, The Netherlands
| | - J M Vink
- Department of Biological Psychology/Netherlands Twin Register, VU University, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
42
|
Ma Y, Wang M, Yuan W, Su K, Li MD. The significant association of Taq1A genotypes in DRD2/ANKK1 with smoking cessation in a large-scale meta-analysis of Caucasian populations. Transl Psychiatry 2015; 5:e686. [PMID: 26624925 PMCID: PMC5068580 DOI: 10.1038/tp.2015.176] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/11/2015] [Accepted: 09/29/2015] [Indexed: 11/23/2022] Open
Abstract
Although a number of studies have analyzed the relation between the DRD2/ANKK1 gene Taq1A polymorphism and smoking cessation, the results remain controversial. The primary objective of the present study was to determine whether this variant indeed has any effect on smoking cessation. The A1-dominant model that considers A1/* (*=A1 or A2) and A2/A2 as two genotypes and compares their frequencies in current and former smokers was applied. A total of 22 studies with 11,075 subjects were included in the meta-analyses. Considering the potential influence of between-study heterogeneity, we conducted stratified meta-analyses with the Comprehensive Meta-Analysis statistical software (version 2.0). Results based on either cross-sectional or longitudinal studies consistently showed a statistically significant association between Taq1A A1/* genotypes and smoking cessation. Further, a more significant association of the variant with smoking cessation was detected when both types of studies were combined. However, there was marginal evidence of heterogeneity among studies (I(2)=33.9%; P=0.06). By excluding other ethnicities and subjects with cancer, the meta-analysis on the basis of 9487 Caucasians demonstrated that Taq1A A1/* genotypes indeed were significantly associated with smoking cessation under both the fixed- and random-effects models (pooled OR 1.22; 95% CI 1.11-1.34; P=3.9 × 10(-5) for both models). No evidence of between-study heterogeneity or publication bias was observed. Thus, we conclude that the polymorphism of Taq1A has an important role in the process of abstaining from smoking, and smokers carrying A2/A2 genotype have a higher likelihood of smoking cessation than those who carry A1/A1 or A1/A2.
Collapse
Affiliation(s)
- Y Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - M Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - W Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - K Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - M D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
- Air Center for Air Pollution and Health, Zhejiang University, Hangzhou, China
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
43
|
Yang J, Wang S, Yang Z, Hodgkinson CA, Iarikova P, Ma JZ, Payne TJ, Goldman D, Li MD. The contribution of rare and common variants in 30 genes to risk nicotine dependence. Mol Psychiatry 2015; 20:1467-78. [PMID: 25450229 PMCID: PMC4452458 DOI: 10.1038/mp.2014.156] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/28/2014] [Accepted: 10/08/2014] [Indexed: 01/17/2023]
Abstract
Genetic and functional studies have revealed that both common and rare variants of several nicotinic acetylcholine receptor subunits are associated with nicotine dependence (ND). In this study, we identified variants in 30 candidate genes including nicotinic receptors in 200 sib pairs selected from the Mid-South Tobacco Family population with equal numbers of African Americans (AAs) and European Americans (EAs). We selected 135 of the rare and common variants and genotyped them in the Mid-South Tobacco Case-Control (MSTCC) population, which consists of 3088 AAs and 1430 EAs. None of the genotyped common variants showed significant association with smoking status (smokers vs non-smokers), Fagerström Test for ND scores or indexed cigarettes per day after Bonferroni correction. Rare variants in NRXN1, CHRNA9, CHRNA2, NTRK2, GABBR2, GRIN3A, DNM1, NRXN2, NRXN3 and ARRB2 were significantly associated with smoking status in the MSTCC AA sample, with weighted sum statistic (WSS) P-values ranging from 2.42 × 10(-3) to 1.31 × 10(-4) after 10(6) phenotype rearrangements. We also observed a significant excess of rare nonsynonymous variants exclusive to EA smokers in NRXN1, CHRNA9, TAS2R38, GRIN3A, DBH, ANKK1/DRD2, NRXN3 and CDH13 with WSS P-values between 3.5 × 10(-5) and 1 × 10(-6). Variants rs142807401 (A432T) and rs139982841 (A452V) in CHRNA9 and variants V132L, V389L, rs34755188 (R480H) and rs75981117 (N549S) in GRIN3A are of particular interest because they are found in both the AA and EA samples. A significant aggregate contribution of rare and common coding variants in CHRNA9 to the risk for ND (SKAT-C: P=0.0012) was detected by applying the combined sum test in MSTCC EAs. Together, our results indicate that rare variants alone or combined with common variants in a subset of 30 biological candidate genes contribute substantially to the risk of ND.
Collapse
Affiliation(s)
- Jiekun Yang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903
| | - Shaolin Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903
| | - Zhongli Yang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903
| | | | | | - Jennie Z. Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville
| | - Thomas J. Payne
- ACT Center for Tobacco Treatment, Education and Research, Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS 39213
| | - David Goldman
- Laboratory of Neurogenetics, NIAAA, NIH; Bethesda, MD 20852
| | - Ming D. Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
44
|
Celorrio D, Muñoz X, Amiano P, Dorronsoro M, Bujanda L, Sánchez MJ, Molina-Montes E, Navarro C, Chirlaque MD, MaríaHuerta J, Ardanaz E, Barricarte A, Rodriguez L, Duell EJ, Hijona E, Herreros-Villanueva M, Sala N, Alfonso-Sánchez MA, de Pancorbo MM. Influence of Dopaminergic System Genetic Variation and Lifestyle Factors on Excessive Alcohol Consumption. Alcohol Alcohol 2015; 51:258-67. [PMID: 26447226 DOI: 10.1093/alcalc/agv114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 07/30/2015] [Indexed: 01/13/2023] Open
Abstract
AIMS To examine the role of genetic and environmental factors in the pathogenesis of alcohol dependence in a Spanish cohort of women and men. METHODS We analyzed the relationship between 56 genetic variants in 7 genes associated with the dopaminergic reward pathway and excessive alcohol consumption. The study sample (N = 1533, of which 746 were women) consisted of 653 heavy consumers and 880 very low consumers from the Spanish subcohort of the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Single nucleotide polymorphisms (SNPs) were genotyped using a customized array. Lifestyle variables were also examined to assess associations between genetic and environmental factors. RESULTS No statistically significant differences were found between cases and controls for the allele frequencies in five genes: TH, SLC18A2, DRD1, DRD3 and COMT. Conversely, some alleles of the 12 SNPs from the DRD2 locus and the 5 from the MAOA locus showed significant associations with excessive alcohol consumption. Namely, rs10891556 (DRD2) proved to be the only SNP positively correlated with excessive alcohol consumption in both sexes. DRD2 rs1800497 and rs877138 were significantly associated in men, whereas DRD2 rs17601612 and rs4936271 and MAOA rs5906898 were associated with excessive alcohol consumption in women. A correspondence analysis provided an overall lifestyle profile of excessive drinkers, who were predominantly men who smoked, had large intakes of meat, small intakes of fruit and vegetables, whose jobs did not require high education levels and who engaged in little physical activity. CONCLUSIONS It has shown the influence of dopaminergic pathway in the genetics of alcohol dependence with differences between men and women and providing a lifestyle profile of excessive drinkers.
Collapse
Affiliation(s)
- David Celorrio
- BIOMICs Research Group, 'Lucio Lascaray' Center for Research and Advanced Studies (CIEA), University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Xavier Muñoz
- Molecular Epidemiology Group, Translational Research Laboratory, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain Unit of Nutrition, and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain
| | - Pilar Amiano
- Public Health Division of Gipuzkoa, Department of Health of the Regional Government of the Basque Country, Donostia, Spain BIODonostia Research Institute, San Sebastián, Spain CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain
| | - Miren Dorronsoro
- Public Health Division of Gipuzkoa, Department of Health of the Regional Government of the Basque Country, Donostia, Spain BIODonostia Research Institute, San Sebastián, Spain CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain
| | - Luis Bujanda
- Departmenet of Gastroenterology, Donostia Hospital-Instituto Biodonostia, University of the Basque Country (UPV/EHU), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), San Sebastian, Spain
| | - María-José Sánchez
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Andalusian School of Public Health, Granada, Spain
| | - Esther Molina-Montes
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Andalusian School of Public Health, Granada, Spain
| | - Carmen Navarro
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Department of Epidemiology, Murcia Health Council, Murcia, Spain
| | - M Dolores Chirlaque
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Department of Epidemiology, Murcia Health Council, Murcia, Spain
| | - José MaríaHuerta
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Department of Epidemiology, Murcia Health Council, Murcia, Spain
| | - Eva Ardanaz
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Public Health Institute of Navarra, Pamplona, Spain
| | - Aurelio Barricarte
- CIBER Epidemiología y Salud Pública CIBERESP, Barcelona, Spain Public Health Institute of Navarra, Pamplona, Spain
| | | | - Eric J Duell
- Unit of Nutrition, and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain
| | - Elizabeth Hijona
- Departmenet of Gastroenterology, Donostia Hospital-Instituto Biodonostia, University of the Basque Country (UPV/EHU), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), San Sebastian, Spain
| | - Marta Herreros-Villanueva
- Departmenet of Gastroenterology, Donostia Hospital-Instituto Biodonostia, University of the Basque Country (UPV/EHU), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), San Sebastian, Spain
| | - Núria Sala
- Molecular Epidemiology Group, Translational Research Laboratory, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain Unit of Nutrition, and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain
| | - Miguel A Alfonso-Sánchez
- BIOMICs Research Group, 'Lucio Lascaray' Center for Research and Advanced Studies (CIEA), University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group, 'Lucio Lascaray' Center for Research and Advanced Studies (CIEA), University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
45
|
MacKillop J, Gray JC, Bidwell LC, Bickel WK, Sheffer CE, McGeary JE. Genetic influences on delay discounting in smokers: examination of a priori candidates and exploration of dopamine-related haplotypes. Psychopharmacology (Berl) 2015; 232. [PMID: 26220612 PMCID: PMC4845660 DOI: 10.1007/s00213-015-4029-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE Delay discounting is a behavioral economic index of impulsivity that reflects a person's relative preference for small immediate rewards versus larger delayed rewards. Elevated delay discounting is robustly linked to addictive disorders and has been increasingly investigated as a viable endophenotype for genetic influences on addiction. OBJECTIVE The aim of this study is to examine associations between delay discounting and two a priori loci, rs4680 in COMT and rs1800497 in ANKK1, and three exploratory haplotypes proximal to rs1800497 in a sample of daily smokers. METHODS Participants were 713 (60.2 % male) daily smokers of European ancestry who completed a delay discounting assessment and provided a DNA sample. RESULTS Significant associations were detected between greater discounting of medium magnitude rewards (~$55) and the G allele of rs4680, as well as the T allele of rs1800497. Exploratory haplotype analyses identified two haplotypes (rs1160467/rs1800497; rs6277/rs1079597) significantly associated with delay discounting rates. However, the rs1160467/rs1800497 haplotype associations appeared to be entirely attributable to variation in rs1800497, suggesting that the association of rs1800497 with discounting is best understood at the individual SNP level. Similarly, the rs6277/rs1079597 haplotype findings suggested that the association was specific to rs1079597. CONCLUSIONS This study provides further evidence that rs4680 and rs1800497 genotypes are significantly associated with delay discounting preferences and does so among smokers for the first time. The study also provides evidence of specificity for the rs1800497 association and identifies a novel locus, rs1079597, as a genetic contributor to higher delay discounting rates.
Collapse
Affiliation(s)
- James MacKillop
- Peter Boris Centre for Addictions Research, Department of Psychiatry and Behavioural Neurosciences, McMaster University/St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada,
| | | | | | | | | | | |
Collapse
|
46
|
Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK Biobank. THE LANCET. RESPIRATORY MEDICINE 2015; 3:769-81. [PMID: 26423011 PMCID: PMC4593935 DOI: 10.1016/s2213-2600(15)00283-0] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Understanding the genetic basis of airflow obstruction and smoking behaviour is key to determining the pathophysiology of chronic obstructive pulmonary disease (COPD). We used UK Biobank data to study the genetic causes of smoking behaviour and lung health. METHODS We sampled individuals of European ancestry from UK Biobank, from the middle and extremes of the forced expiratory volume in 1 s (FEV1) distribution among heavy smokers (mean 35 pack-years) and never smokers. We developed a custom array for UK Biobank to provide optimum genome-wide coverage of common and low-frequency variants, dense coverage of genomic regions already implicated in lung health and disease, and to assay rare coding variants relevant to the UK population. We investigated whether there were shared genetic causes between different phenotypes defined by extremes of FEV1. We also looked for novel variants associated with extremes of FEV1 and smoking behaviour and assessed regions of the genome that had already shown evidence for a role in lung health and disease. We set genome-wide significance at p<5 × 10(-8). FINDINGS UK Biobank participants were recruited from March 15, 2006, to July 7, 2010. Sample selection for the UK BiLEVE study started on Nov 22, 2012, and was completed on Dec 20, 2012. We selected 50,008 unique samples: 10,002 individuals with low FEV1, 10,000 with average FEV1, and 5002 with high FEV1 from each of the heavy smoker and never smoker groups. We noted a substantial sharing of genetic causes of low FEV1 between heavy smokers and never smokers (p=2.29 × 10(-16)) and between individuals with and without doctor-diagnosed asthma (p=6.06 × 10(-11)). We discovered six novel genome-wide significant signals of association with extremes of FEV1, including signals at four novel loci (KANSL1, TSEN54, TET2, and RBM19/TBX5) and independent signals at two previously reported loci (NPNT and HLA-DQB1/HLA-DQA2). These variants also showed association with COPD, including in individuals with no history of smoking. The number of copies of a 150 kb region containing the 5' end of KANSL1, a gene that is important for epigenetic gene regulation, was associated with extremes of FEV1. We also discovered five new genome-wide significant signals for smoking behaviour, including a variant in NCAM1 (chromosome 11) and a variant on chromosome 2 (between TEX41 and PABPC1P2) that has a trans effect on expression of NCAM1 in brain tissue. INTERPRETATION By sampling from the extremes of the lung function distribution in UK Biobank, we identified novel genetic causes of lung function and smoking behaviour. These results provide new insight into the specific mechanisms underlying airflow obstruction, COPD, and tobacco addiction, and show substantial shared genetic architecture underlying airflow obstruction across individuals, irrespective of smoking behaviour and other airway disease. FUNDING Medical Research Council.
Collapse
|
47
|
Bidwell LC, McGeary JE, Gray JC, Palmer RHC, Knopik VS, MacKillop J. An initial investigation of associations between dopamine-linked genetic variation and smoking motives in African Americans. Pharmacol Biochem Behav 2015; 138:104-10. [PMID: 26410615 DOI: 10.1016/j.pbb.2015.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/05/2015] [Accepted: 09/24/2015] [Indexed: 11/25/2022]
Abstract
Nicotine dependence (ND) is a heterogeneous phenotype with complex genetic influences that may vary across ethnicities. The use of intermediate phenotypes may clarify genetic influences and reveal specific etiological pathways. Prior work in European Americans has found that the four Primary Dependence Motives (PDM) subscales (Automaticity, Craving, Loss of Control, and Tolerance) of the Wisconsin Inventory of Smoking Motives represent core features of nicotine dependence and are promising intermediate phenotypes for understanding genetic pathways to ND. However, no studies have examined PDM as an intermediate phenotype in African American smokers, an ethnic population that displays unique patterns of smoking and genetic variation. In the current study, 268 African American daily smokers completed a phenotypic assessment and provided a sample of DNA. Associations among haplotypes in the NCAM1-TTC12-ANKK1-DRD2 gene cluster, a dopamine-related gene region associated with ND, PDM intermediate phenotypes, and ND were examined. Dopamine-related genetic variation in the DBH and COMT genes was also considered on an exploratory basis. Mediational analysis was used to test the indirect pathway from genetic variation to smoking motives to nicotine dependence. NCAM1-TTC12-ANKK1-DRD2 region variation was significantly associated with the Automaticity subscale and, further, Automaticity significantly mediated associations among NCAM1-TTC12-ANKK1-DRD2 cluster variants and ND. DBH was also significantly associated with Automaticity, Craving, and Tolerance; Automaticity and Tolerance also served as mediators of the DBH-ND relationship. These results suggest that PDM, Automaticity in particular, may be a viable intermediate phenotype for understanding dopamine-related genetic influences on ND in African American smokers. Findings support a model in which putatively dopaminergic variants exert influence on ND through an effect on patterns of automatic routinized smoking.
Collapse
Affiliation(s)
- L C Bidwell
- Institute of Cognitive Science, University of Colorado at Boulder, Boulder, CO 80304, United States; Division of Behavioral Genetics, Rhode Island Hospital, 1 Hoppin St., Providence, RI 02903, United States; Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI 02903, United States.
| | - J E McGeary
- Providence Veterans Affairs Medical Center, 830 Chalkstone Avenue, Building 35, Providence, RI 02908, United States; Division of Behavioral Genetics, Rhode Island Hospital, 1 Hoppin St., Providence, RI 02903, United States; Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI 02903, United States
| | - J C Gray
- Department of Psychology, 100 Hooper St., University of Georgia, Athens, GA 30602, United States
| | - R H C Palmer
- Division of Behavioral Genetics, Rhode Island Hospital, 1 Hoppin St., Providence, RI 02903, United States; Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI 02903, United States
| | - V S Knopik
- Division of Behavioral Genetics, Rhode Island Hospital, 1 Hoppin St., Providence, RI 02903, United States; Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI 02903, United States
| | - J MacKillop
- Peter Boris Centre for Addictions Research, McMaster University/St. Joseph's Healthcare Hamilton, 100 West 5th At., Hamilton, ON L8N 3K7, Canada
| |
Collapse
|
48
|
Mota NR, Rovaris DL, Kappel DB, Picon FA, Vitola ES, Salgado CAI, Karam RG, Rohde LA, Grevet EH, Bau CHD. NCAM1-TTC12-ANKK1-DRD2 gene cluster and the clinical and genetic heterogeneity of adults with ADHD. Am J Med Genet B Neuropsychiatr Genet 2015; 168:433-444. [PMID: 25989041 DOI: 10.1002/ajmg.b.32317] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 04/07/2015] [Indexed: 12/23/2022]
Abstract
Dysfunctions of the dopaminergic system have been implicated on the etiology of Attention Deficit/Hyperactivity Disorder (ADHD). Meta-analyses addressing the association of the dopamine receptor D2 (DRD2) gene and ADHD were inconclusive due to excessive heterogeneity across studies. Both the great phenotypic heterogeneity of ADHD and the complexity of the genomic region where DRD2 is located could contribute to the inconsistent findings. Most previous DRD2 studies focused on the well-known Taq1A (rs1800497) SNP, which is actually placed in a neighbor gene (ANKK1). These two genes, together with NCAM1 and TTC12, form the NTAD gene cluster on Chr11q22-23. In order to address the reasons for the high heterogeneity previously reported on DRD2 effects on ADHD, this study investigates the role of NTAD variants on ADHD susceptibility in adults and on the modulation of comorbidity and personality profiles in these patients. Functional polymorphisms from NTAD were analyzed, both individually and in haplotypes, on a sample of 520 adults with ADHD and 630 non-ADHD controls. No direct association of NTAD variants with ADHD susceptibility itself was observed. However, different NTAD polymorphisms and haplotypes were associated to various phenotypes relevant to the clinical heterogeneity of ADHD, including Major Depressive Disorder, Generalized Anxiety Disorder, and Harm Avoidance and Persistence temperament scores. Therefore, these findings represent a possible explanation for the multiple conflicting findings regarding polymorphisms in this genomic region in psychiatry. The NTAD cluster may comprise a variety of independent molecular influences on various brain and behavior characteristics eventually associated with ADHD comorbidities and personality traits. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nina R Mota
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Diego L Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Djenifer B Kappel
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Felipe A Picon
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Eduardo S Vitola
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Carlos A I Salgado
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Rafael G Karam
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| | - Luis A Rohde
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eugenio H Grevet
- ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil.,Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Claiton H D Bau
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,ADHD Outpatient Program-Adult Division, Hospital de Clínicas, de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
49
|
Villalba K, Devieux JG, Rosenberg R, Cadet JL. DRD2 and DRD4 genes related to cognitive deficits in HIV-infected adults who abuse alcohol. Behav Brain Funct 2015; 11:25. [PMID: 26307064 PMCID: PMC4549947 DOI: 10.1186/s12993-015-0072-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/17/2015] [Indexed: 11/23/2022] Open
Abstract
Background HIV-infected individuals continue to
experience neurocognitive deterioration despite virologically successful treatments. The causes of neurocognitive impairment are still unclear. However, several factors have been suggested including the role of genetics. There is evidence suggesting that neurocognitive impairment is heritable and individual differences in cognition are strongly driven by genetic variations. The contribution of genetic variants affecting the metabolism and activity of dopamine may influence these individual differences. Methods The present study explored the relationship between two candidate genes (DRD4 and DRD2) and neurocognitive performance in HIV-infected adults. A total of 267 HIV-infected adults were genotyped for polymorphisms, DRD4 48 bp-variable number tandem repeat (VNTR), DRD2 rs6277 and ANKK1 rs1800497. The Short Category (SCT), Color Trail (CTT) and Rey-Osterrieth Complex Figure Tests (ROCT) were used to measure executive function and memory. Results Results showed significant associations with the SNP rs6277 and impaired executive function (odds ratio = 3.3, 95 % CI 1.2–2.6; p = 0.004) and cognitive flexibility (odds ratio = 1.6, 95 % CI 2.0–5.7; p = 0.001). The results were further stratified by race and sex and significant results were seen in males (odds ratio = 3.5, 95 % CI 1.5–5.5; p = 0.008) and in African Americans (odds ratio = 3.1, 95 % CI 2.3–3.5; p = 0.01). Also, DRD4 VNTR 7-allele was significantly associated with executive dysfunction. Conclusion The study shows that genetically determined differences in the SNP rs6277 DRD2 gene and DRD4 48 bp VNTR may be risk factors for deficits in executive function and cognitive flexibility.
Collapse
Affiliation(s)
- Karina Villalba
- Department of Health Promotion and Disease Prevention, Robert Stempel College of Public Health and Social Work, Florida International University, Biscayne Bay Campus, 3000 N.E, 151 Street ACI #260, North Miami, FL, 33181, USA.
| | - Jessy G Devieux
- Department of Health Promotion and Disease Prevention, Robert Stempel College of Public Health and Social Work, Florida International University, Biscayne Bay Campus, 3000 N.E, 151 Street ACI #260, North Miami, FL, 33181, USA
| | - Rhonda Rosenberg
- Department of Health Promotion and Disease Prevention, Robert Stempel College of Public Health and Social Work, Florida International University, Biscayne Bay Campus, 3000 N.E, 151 Street ACI #260, North Miami, FL, 33181, USA
| | - Jean Lud Cadet
- NIDA Intramural Program, Molecular Neuropsychiatry Research Branch, Baltimore, MD, USA
| |
Collapse
|
50
|
Hart AB, Kranzler HR. Alcohol Dependence Genetics: Lessons Learned From Genome-Wide Association Studies (GWAS) and Post-GWAS Analyses. Alcohol Clin Exp Res 2015; 39:1312-27. [PMID: 26110981 PMCID: PMC4515198 DOI: 10.1111/acer.12792] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/26/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alcohol dependence (AD) is a complex psychiatric disorder and a significant public health problem. Twin and family-based studies have consistently estimated its heritability to be approximately 50%, and many studies have sought to identify specific genetic variants associated with susceptibility to AD. These studies have been primarily linkage or candidate gene based and have been mostly unsuccessful in identifying replicable risk loci. Genome-wide association studies (GWAS) have improved the detection of specific loci associated with complex traits, including AD. However, findings from GWAS explain only a small proportion of phenotypic variance, and alternative methods have been proposed to investigate the associations that do not meet strict genome-wide significance criteria. METHODS This review summarizes all published AD GWAS and post-GWAS analyses that have sought to exploit GWAS data to identify AD-associated loci. RESULTS Findings from AD GWAS have been largely inconsistent, with the exception of variants encoding the alcohol-metabolizing enzymes. Analyses of GWAS data that go beyond standard association testing have demonstrated the polygenic nature of AD and the large contribution of common variants to risk, nominating novel genes and pathways for AD susceptibility. CONCLUSIONS Findings from AD GWAS and post-GWAS analyses have greatly increased our understanding of the genetic etiology of AD. However, it is clear that larger samples will be necessary to detect loci in addition to those that encode alcohol-metabolizing enzymes, which may only be possible through consortium-based efforts. Post-GWAS approaches to studying the genetic influences on AD are increasingly common and could greatly increase our knowledge of both the genetic architecture of AD and the specific genes and pathways that influence risk.
Collapse
Affiliation(s)
- Amy B. Hart
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- VISN 4 MIRECC, Philadelphia VAMC, Philadelphia, PA 19104, USA
| |
Collapse
|