1
|
Song H, Yang P, Zhang X, Tao R, Zuo L, Liu W, Fu J, Kong Z, Tang R, Wu S, Pang L, Zhang X. Atypical effective connectivity from the frontal cortex to striatum in alcohol use disorder. Transl Psychiatry 2024; 14:381. [PMID: 39294121 PMCID: PMC11411137 DOI: 10.1038/s41398-024-03083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
Alcohol use disorder (AUD) is a profound psychiatric condition marked by disrupted connectivity among distributed brain regions, indicating impaired functional integration. Previous connectome studies utilizing functional magnetic resonance imaging (fMRI) have predominantly focused on undirected functional connectivity, while the specific alterations in directed effective connectivity (EC) associated with AUD remain unclear. To address this issue, this study utilized multivariate pattern analysis (MVPA) and spectral dynamic causal modeling (DCM). We recruited 32 abstinent men with AUD and 30 healthy controls (HCs) men, and collected their resting-state fMRI data. A regional homogeneity (ReHo)-based MVPA method was employed to classify AUD and HC groups, as well as predict the severity of addiction in AUD individuals. The most informative brain regions identified by the MVPA were further investigated using spectral DCM. Our results indicated that the ReHo-based support vector classification (SVC) exhibits the highest accuracy in distinguishing individuals with AUD from HCs (classification accuracy: 98.57%). Additionally, our results demonstrated that ReHo-based support vector regression (SVR) could be utilized to predict the addiction severity (alcohol use disorders identification test, AUDIT, R2 = 0.38; Michigan alcoholism screening test, MAST, R2 = 0.29) of patients with AUD. The most informative brain regions for the prediction include left pre-SMA, right dACC, right LOFC, right putamen, and right NACC. These findings were validated in an independent data set (35 patients with AUD and 36 HCs, Classification accuracy: 91.67%; AUDIT, R2 = 0.17; MAST, R2 = 0.20). The results of spectral DCM analysis indicated that individuals with AUD exhibited decreased EC from the left pre-SMA to the right putamen, from the right dACC to the right putamen, and from the right LOFC to the right NACC compared to HCs. Moreover, the EC strength from the right NACC to left pre-SMA and from the right dACC to right putamen mediated the relationship between addiction severity (MAST scores) and behavioral measures (impulsive and compulsive scores). These findings provide crucial evidence for the underlying mechanism of impaired self-control, risk assessment, and impulsive and compulsive alcohol consumption in individuals with AUD, providing novel causal insights into both diagnosis and treatment.
Collapse
Affiliation(s)
- Hongwen Song
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Philosophy and Social Science of Anhui Province on Adolescent Mental Health and Crisis Intelligence Intervention, Hefei Normal University, Hefei, China
- The Institute of Linguistics and Applied Linguistics, Anhui Jianzhu University, Hefei, China
| | - Ping Yang
- Department of Psychology, School of Humanities and Social Science, University of Science and Technology of China, Hefei, China
| | - Xinyue Zhang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Tao
- Department of Substance-Related Disorders, Hefei Fourth People's Hospital, Hefei, China
| | - Lin Zuo
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Weili Liu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaxin Fu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhuo Kong
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Tang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Siyu Wu
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Liangjun Pang
- Department of Substance-Related Disorders, Hefei Fourth People's Hospital, Hefei, China.
| | - Xiaochu Zhang
- Department of Radiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale and School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Psychology, School of Humanities and Social Science, University of Science and Technology of China, Hefei, China.
- School of Mental Health, Bengbu Medical College, Bengbu, China.
- Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China.
- Institute of Health and Medicine, Hefei Comprehensive Science Center, Hefei, China.
| |
Collapse
|
2
|
Trang KB, Chesi A, Toikumo S, Pippin JA, Pahl MC, O’Brien JM, Amundadottir LT, Brown KM, Yang W, Welles J, Santoleri D, Titchenell PM, Seale P, Zemel BS, Wagley Y, Hankenson KD, Kaestner KH, Anderson SA, Kayser MS, Wells AD, Kranzler HR, Kember RL, Grant SF. Shared and unique 3D genomic features of substance use disorders across multiple cell types. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.18.24310649. [PMID: 39072016 PMCID: PMC11275669 DOI: 10.1101/2024.07.18.24310649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Recent genome-wide association studies (GWAS) have revealed shared genetic components among alcohol, opioid, tobacco and cannabis use disorders. However, the extent of the underlying shared causal variants and effector genes, along with their cellular context, remain unclear. We leveraged our existing 3D genomic datasets comprising high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq and RNA-seq across >50 diverse human cell types to focus on genomic regions that coincide with GWAS loci. Using stratified LD regression, we determined the proportion of genomewide SNP heritability attributable to the features assayed across our cell types by integrating recent GWAS summary statistics for the relevant traits: alcohol use disorder (AUD), tobacco use disorder (TUD), opioid use disorder (OUD) and cannabis use disorder (CanUD). Statistically significant enrichments (P<0.05) were observed in 14 specific cell types, with heritability reaching 9.2-fold for iPSC-derived cortical neurons and neural progenitors, confirming that they are crucial cell types for further functional exploration. Additionally, several pancreatic cell types, notably pancreatic beta cells, showed enrichment for TUD, with heritability enrichments up to 4.8-fold, suggesting genomic overlap with metabolic processes. Further investigation revealed significant positive genetic correlations between T2D with both TUD and CanUD (FDR<0.05) and a significant negative genetic correlation with AUD. Interestingly, after partitioning the heritability for each cell type's cis-regulatory elements, the correlation between T2D and TUD for pancreatic beta cells was greater (r=0.2) than the global genetic correlation value. Our study provides new genomic insights into substance use disorders and implicates cell types where functional follow-up studies could reveal causal variant-gene mechanisms underpinning these disorders.
Collapse
Affiliation(s)
- Khanh B. Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joan M. O’Brien
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Disease, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M. Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wenli Yang
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaclyn Welles
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominic Santoleri
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yadav Wagley
- Department of Orthopedic Surgery, University of Michigan Medical School Ann Arbor, MI, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School Ann Arbor, MI, USA
| | - Klaus H. Kaestner
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew S. Kayser
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel L. Kember
- Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
3
|
Lai D, Zhang M, Green N, Abreu M, Schwantes-An TH, Parker C, Zhang S, Jin F, Sun A, Zhang P, Edenberg H, Liu Y, Foroud T. Genome-wide meta-analyses of cross substance use disorders in European, African, and Latino ancestry populations. RESEARCH SQUARE 2024:rs.3.rs-3955955. [PMID: 39070649 PMCID: PMC11275984 DOI: 10.21203/rs.3.rs-3955955/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Genetic risks for substance use disorders (SUDs) are due to both SUD-specific and SUD-shared genes. We performed the largest multivariate analyses to date to search for SUD-shared genes using samples of European (EA), African (AA), and Latino (LA) ancestries. By focusing on variants having cross-SUD and cross-ancestry concordant effects, we identified 45 loci. Through gene-based analyses, gene mapping, and gene prioritization, we identified 250 SUD-shared genes. These genes are highly expressed in amygdala, cortex, hippocampus, hypothalamus, and thalamus, primarily in neuronal cells. Cross-SUD concordant variants explained ~ 50% of the heritability of each SUD in EA. The top 5% individuals having the highest polygenic scores were approximately twice as likely to have SUDs as others in EA and LA. Polygenic scores had higher predictability in females than in males in EA. Using real-world data, we identified five drugs targeting identified SUD-shared genes that may be repurposed to treat SUDs.
Collapse
Affiliation(s)
- Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | | | | | | | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | | | | | | | - Anna Sun
- Indiana University School of Medicine
| | | | | | | | | |
Collapse
|
4
|
Miller AP, Bogdan R, Agrawal A, Hatoum AS. Generalized genetic liability to substance use disorders. J Clin Invest 2024; 134:e172881. [PMID: 38828723 PMCID: PMC11142744 DOI: 10.1172/jci172881] [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] [Indexed: 06/05/2024] Open
Abstract
Lifetime and temporal co-occurrence of substance use disorders (SUDs) is common and compared with individual SUDs is characterized by greater severity, additional psychiatric comorbidities, and worse outcomes. Here, we review evidence for the role of generalized genetic liability to various SUDs. Coaggregation of SUDs has familial contributions, with twin studies suggesting a strong contribution of additive genetic influences undergirding use disorders for a variety of substances (including alcohol, nicotine, cannabis, and others). GWAS have documented similarly large genetic correlations between alcohol, cannabis, and opioid use disorders. Extending these findings, recent studies have identified multiple genomic loci that contribute to common risk for these SUDs and problematic tobacco use, implicating dopaminergic regulatory and neuronal development mechanisms in the pathophysiology of generalized SUD genetic liability, with certain signals demonstrating cross-species and translational validity. Overlap with genetic signals for other externalizing behaviors, while substantial, does not explain the entirety of the generalized genetic signal for SUD. Polygenic scores (PGS) derived from the generalized genetic liability to SUDs outperform PGS for individual SUDs in prediction of serious mental health and medical comorbidities. Going forward, it will be important to further elucidate the etiology of generalized SUD genetic liability by incorporating additional SUDs, evaluating clinical presentation across the lifespan, and increasing the granularity of investigation (e.g., specific transdiagnostic criteria) to ultimately improve the nosology, prevention, and treatment of SUDs.
Collapse
Affiliation(s)
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Alexander S. Hatoum
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Kendler KS, Ohlsson H, Sundquist J, Sundquist K. The predictive effect of family genetic risk scores as an indirect measure of causal effects of one disorder on another. Psychol Med 2024; 54:1867-1875. [PMID: 38314515 DOI: 10.1017/s0033291723003847] [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] [Indexed: 02/06/2024]
Abstract
BACKGROUND One potential cause of comorbidity is the direct causal effect of one disorder - A - on risk for subsequent onset of disorder B. Could genetic risk scores be utilized to test for such an effect? If disorder A causally impacts on risk for disorder B, then genetic risk for disorder A should be lower in cases of disorder A with v. without a prior onset of B. METHODS In all individuals (n = 905 736) born in Sweden from 1980 to 1990, from six psychiatric and drug use disorders (major depression, anxiety disorders, alcohol use disorder, drug use disorder, bipolar disorder, and schizophrenia), we formed 14 pairs of disorders A and B. In these pairs, we compared, using Cox proportional hazards models, the predictive effect of the familial-genetic risk score (FGRS) for disorder B in those who had v. had not had a prior onset of disorder A. RESULTS In all pairs, the impact of the FGRS for disorder B was significantly stronger in cases without v. with a prior history of disorder A. These effects were similar across sex, stable across levels of FGRS and not likely due to clinician bias. In many of our disorder pairs, previous clinical studies suggest a mechanism for a causal effect of disorder A on B. CONCLUSIONS Our findings provide indirect evidence that the occurrence of one psychiatric or substance use disorder often has a causal effect on risk for subsequent disorders. This mechanism may substantially contribute to the widespread comorbidity among psychiatric conditions.
Collapse
Affiliation(s)
- Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Henrik Ohlsson
- Center for Primary Health Care Research, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
- University Clinic Primary Care Skåne, Region Skåne, Sweden
| | - Kristina Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
- University Clinic Primary Care Skåne, Region Skåne, Sweden
| |
Collapse
|
6
|
Kendler KS, Abrahamsson L, Sundquist J, Sundquist K. Pattern of Risks for Psychiatric and Substance Use Disorders in the Offspring of Parents With Alcohol Use Disorder. Am J Psychiatry 2024; 181:322-329. [PMID: 38419493 DOI: 10.1176/appi.ajp.20230376] [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] [Indexed: 03/02/2024]
Abstract
OBJECTIVE The authors sought to clarify the components of the familial liability to alcohol use disorder (AUD) by examining parent-offspring transmission in a large Swedish population sample. METHODS To this end, 1,244,516 offspring in intact families with a mean age at follow-up of 37.7 years (SD=6.8) were examined. Hazard ratios for offspring of parents with AUD were calculated using Cox models for risk of five disorders assessed from Swedish medical and criminal registries: AUD, drug use disorders, attention deficit hyperactivity disorder, major depression, and anxiety disorders. RESULTS The hazard ratio for the offspring was highest for AUD (hazard ratio=2.36), followed by drug use disorder (hazard ratio=2.04), attention deficit hyperactivity disorder (hazard ratio=1.82), major depression (hazard ratio=1.43), and anxiety disorder (hazard ratio=1.43). The risks for AUD were statistically indistinguishable between the children having mothers with AUD compared with those having fathers with AUD and between sons and daughters of a parent with AUD. All risks for offspring having two parents with AUD were higher than those having one parent with AUD, but the increase with two parents with AUD was greatest for AUD, followed by drug use disorder and attention deficit hyperactivity disorder. Age at AUD onset of the parents predicted risk among the offspring more strongly for AUD and drug use disorder, followed by attention deficit hyperactivity disorder, and then major depression and anxiety disorders. Number of recurrences of the parents with AUD predicted risks for all disorders equally. The risk pattern of disorders for the offspring of not-lived-with fathers with AUD was similar to that in the main analysis of intact families. No evidence was found for sex-specific transmission of AUD or a familial female protective effect. CONCLUSIONS Familial and likely genetic liability to AUD has three components: a nonspecific risk of common internalizing and externalizing disorders, a moderately specific risk of externalizing disorders, and a highly specific risk of AUD.
Collapse
Affiliation(s)
- Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics and Department of Psychiatry, Virginia Commonwealth University, Richmond (Kendler); Center for Primary Health Care Research, Lund University, Malmö, Sweden (Abrahamsson, Jan Sundquist, Kristina Sundquist)
| | - Linda Abrahamsson
- Virginia Institute for Psychiatric and Behavioral Genetics and Department of Psychiatry, Virginia Commonwealth University, Richmond (Kendler); Center for Primary Health Care Research, Lund University, Malmö, Sweden (Abrahamsson, Jan Sundquist, Kristina Sundquist)
| | - Jan Sundquist
- Virginia Institute for Psychiatric and Behavioral Genetics and Department of Psychiatry, Virginia Commonwealth University, Richmond (Kendler); Center for Primary Health Care Research, Lund University, Malmö, Sweden (Abrahamsson, Jan Sundquist, Kristina Sundquist)
| | - Kristina Sundquist
- Virginia Institute for Psychiatric and Behavioral Genetics and Department of Psychiatry, Virginia Commonwealth University, Richmond (Kendler); Center for Primary Health Care Research, Lund University, Malmö, Sweden (Abrahamsson, Jan Sundquist, Kristina Sundquist)
| |
Collapse
|
7
|
Miech R, Heeringa SG, Molinaro S, Benedetti E. Cannabis reduction among adolescents as spillover from successful tobacco control. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2024; 124:104315. [PMID: 38183859 PMCID: PMC10939814 DOI: 10.1016/j.drugpo.2023.104315] [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: 08/27/2023] [Revised: 10/27/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND National programs that reduce adolescent cannabis use warrant renewed attention in light of current discussions to reform cannabis legislation, including the possibility of legalization for recreational use. This study measures the size of a decrease in a country's prevalence of adolescent cannabis use that accompanies a decrease in its prevalence of adolescents who had ever smoked a cigarette. METHODS Data are from the European School Survey Project on Alcohol and Other Drugs (ESPAD), which is a collaborative effort of more than 40 European countries to surveil adolescent substance use. This study uses data from the seven survey administrations in 1995, 1999, 2003, 2007, 2011, 2015, and 2019. The main analysis is a fixed-effect regression analysis of country-level, four-year changes in adolescent lifetime cannabis use prevalence on country-level, four-year changes in adolescent lifetime cigarette use prevalence. RESULTS Decreases in the national prevalence of adolescents who had ever smoked a cigarette were accompanied by decreases half as large in national prevalence of adolescent lifetime cannabis use. CONCLUSION For European countries considering the legalization of adult recreational cannabis use, tobacco control can offer a tool to help counter potential increases in cannabis use among adolescents.
Collapse
Affiliation(s)
| | | | - Sabrina Molinaro
- Institute of Clinical Physiology, IFC, at the National Research Council of Italy - CNR, Via Moruzzi 1, 56124 Pisa, IT, Italy
| | - Elisa Benedetti
- Institute of Clinical Physiology, IFC, at the National Research Council of Italy - CNR, Via Moruzzi 1, 56124 Pisa, IT, Italy
| |
Collapse
|
8
|
Vanyukov M. The misnomer of substance use "stigma": Beneficial disapproval should not be conflated with mistreatment of users. ADDICTION RESEARCH & THEORY 2023; 32:101-103. [PMID: 38523740 PMCID: PMC10957147 DOI: 10.1080/16066359.2023.2283574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 03/26/2024]
|
9
|
Wijekumar PJ, Ranadeva NDK, Jayamaha AR, Herath HMNDM, Noorden N, Fernando SSN. A novel tetra-primer ARMS-PCR for genotyping of the OPRM1 gene rs1799971 variant associated with opioid use disorders. BMC Res Notes 2023; 16:333. [PMID: 37964305 PMCID: PMC10648702 DOI: 10.1186/s13104-023-06578-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
OBJECTIVES A SNV is a single nucleotide change that can occur at any point in the genome. SNVs are the most common genetic variants that occur in the human genome, and a number of SNVs have been found to be associated with human traits and disease. Researchers genotype SNVs using TaqMan technology, DNA microarray, MALDI-TOF mass spectrometry, and automated sequencing, which are expensive and time-consuming. The OPRM1 gene rs1799971 (A118G) has been identified for its association with Opioid use disorder (OUD). The present study focused on developing a single step identification test using Tetra-Primer Amplification Refractory Mutation System-PCR (T-ARMS-PCR) to detect the presence of SNV OPRM1 rs1799971 (A118G). This study was performed to optimize the protocol for the designed four primers and validate it using a total of 52 buccal samples from volunteers who are currently under rehabilitation for the drug abuse disorder. RESULTS Utilizing 52 DNA samples, a novel T-ARMS-PCR assay was successfully developed, tested, and validated. The products of the T-ARMS PCR for rs1799971 contained 395 bp as the control band, 186 bp as G allele (variant) and 257 bp as A allele (wild type), which were observed in the gel image. The genotype frequencies for the OPRM1 gene rs1799971 (A118G) were 44% (22/52) of homozygous variant type (GG), 28.9% (15/52) of homozygous wild type (AA) and 28.9% (15/22) of heterozygous (AG). The G allele frequency was 56.7% and A allele frequency was 43.3%.
Collapse
Affiliation(s)
| | | | | | | | | | - S S N Fernando
- Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Jayewardenepura, Sri Lanka
| |
Collapse
|
10
|
Mahyoub MA, Al-Qurmoti S, Rai AA, Abbas M, Jebril M, Alnaggar M, He S. Adverse physiological effects of smoking cessation on the gastrointestinal tract: A review. Medicine (Baltimore) 2023; 102:e35124. [PMID: 37747027 PMCID: PMC10519547 DOI: 10.1097/md.0000000000035124] [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: 05/01/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Smoking cessation is known to have numerous health benefits, but it can also induce adverse physiological effects, including those affecting the gastrointestinal tract (GIT). Understanding the adverse physiological effects of smoking cessation on the GIT is critical for healthcare professionals and smokers attempting to quit, as it enables them to anticipate and manage potential challenges during the smoking cessation process. Although the detrimental effects of smoking on the GIT have been well established, there is a gap in the literature regarding the specific physiological reactions that may occur upon smoking cessation. This mini-review summarizes the current literature on the predisposing factors, pathophysiology, clinical presentation, and treatment options for adverse physiological effects of smoking cessation on the GIT. We aimed to raise awareness among busy clinical professionals about these adverse effects, empowering them to effectively support individuals striving to quit smoking and maintain their cessation. By consolidating the existing knowledge in this field, this review offers practical implications for smokers, healthcare providers, and policymakers to optimize smoking cessation interventions and support strategies to improve health outcomes.
Collapse
Affiliation(s)
- Mueataz A. Mahyoub
- Department of Gastroenterology, Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
- Department of Gastroenterology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Clinical Medical Research Center for Digestive Diseases (Oncology) of Shaanxi Province, Xi’an, China
| | - Sarah Al-Qurmoti
- Department of Cleft Palate-Craniofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | | | - Mustafa Abbas
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Majed Jebril
- College of Health Sciences, Department of Laboratory Medical Sciences, The Islamic University of Gaza, Gaza, Palestine
| | - Mohammed Alnaggar
- Department of Internal Medicine, Clinic Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
- Department of Oncology, South Hubei Cancer Hospital, Xianning, Hubei, China
| | - Shuixiang He
- Department of Gastroenterology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Clinical Medical Research Center for Digestive Diseases (Oncology) of Shaanxi Province, Xi’an, China
| |
Collapse
|
11
|
Polak K, Haug NA, Dillon P, Svikis DS. Substance Use Disorders in Women. Psychiatr Clin North Am 2023; 46:487-503. [PMID: 37500246 DOI: 10.1016/j.psc.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Substance use disorder (SUD) is among the leading causes of premature morbidity and mortality and imposes significant health, economic, and social burdens. Gender differences have been found in the development, course, and treatment of SUD, with women at increased risk for physiologic and psychosocial consequences compared with men. Reasons for these differences are multifold and include biological, genetic, environmental, and behavioral factors. This article discusses SUD among women, emphasizing clinical considerations for care. Specific topics include epidemiology, sex and gender differences, common comorbidities, screening, diagnosis, treatment, pregnancy, and sociocultural factors.
Collapse
Affiliation(s)
- Kathryn Polak
- Department of Psychiatry, Virginia Commonwealth University, 806 West Franklin Street, PO Box 842018, Richmond, VA 23284, USA.
| | - Nancy A Haug
- Department of Psychology, Palo Alto University, 1791 Arastradero Road, Palo Alto, CA 94304, USA
| | - Pamela Dillon
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, 1200 East Clay Street, Richmond, VA 23298, USA
| | - Dace S Svikis
- Department of Psychology, Institute for Women's Health, Virginia Commonwealth University, 806 West Franklin Street, PO Box 842018, Richmond, VA 23284, USA
| |
Collapse
|
12
|
Khan AH, Bagley JR, LaPierre N, Gonzalez-Figueroa C, Spencer TC, Choudhury M, Xiao X, Eskin E, Jentsch JD, Smith DJ. Genetic pathways regulating the longitudinal acquisition of cocaine self-administration in a panel of inbred and recombinant inbred mice. Cell Rep 2023; 42:112856. [PMID: 37481717 PMCID: PMC10530068 DOI: 10.1016/j.celrep.2023.112856] [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: 11/16/2022] [Revised: 06/06/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023] Open
Abstract
To identify addiction genes, we evaluate intravenous self-administration of cocaine or saline in 84 inbred and recombinant inbred mouse strains over 10 days. We integrate the behavior data with brain RNA-seq data from 41 strains. The self-administration of cocaine and that of saline are genetically distinct. We maximize power to map loci for cocaine intake by using a linear mixed model to account for this longitudinal phenotype while correcting for population structure. A total of 15 unique significant loci are identified in the genome-wide association study. A transcriptome-wide association study highlights the Trpv2 ion channel as a key locus for cocaine self-administration as well as identifying 17 additional genes, including Arhgef26, Slc18b1, and Slco5a1. We find numerous instances where alternate splice site selection or RNA editing altered transcript abundance. Our work emphasizes the importance of Trpv2, an ionotropic cannabinoid receptor, for the response to cocaine.
Collapse
Affiliation(s)
- Arshad H Khan
- Department of Molecular and Medical Pharmacology, Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Nathan LaPierre
- Department of Computer Science, UCLA, Los Angeles, CA 90095, USA
| | | | - Tadeo C Spencer
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Mudra Choudhury
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Eleazar Eskin
- Department of Computational Medicine, UCLA, Los Angeles, CA 90095, USA
| | - James D Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Desmond J Smith
- Department of Molecular and Medical Pharmacology, Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
13
|
Kendler KS, Abrahamsson L, Ohlsson H, Sundquist J, Sundquist K. Cross-generational transmission of genetic risk for alcohol and drug use disorders: the impact of substance availability on the specificity of genetic risk. Psychol Med 2023; 53:5109-5118. [PMID: 35993335 PMCID: PMC10016433 DOI: 10.1017/s0033291722002549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Among individuals with alcohol use disorder (AUD) and drug use disorder (DUD), is their genetic liability and its specificity moderated by substance availability? METHODS Offspring (born 1960-1995) and their biological parents from three family types [not-lived-with (NLW) biological father, mother and adoptive] and their AUD and DUD diagnoses were ascertained from Swedish national registers. Parent-offspring resemblance was calculated by tetrachoric correlation. RESULTS In Swedes born from 1960 to 1995, prevalence rates of AUD were stable while DUD rates increased substantially. Best-estimate tetrachoric correlations (±95% confidence intervals) between AUD in biological parents and AUD and DUD in their offspring were, respectively, +0.19 (0.18-0.20) and +0.18 (0.17-0.20). Parallel results from DUD in parents to AUD and DUD in children were +0.12 (0.10-0.13) and +0.27 (0.26-0.28). When divided into older and younger cohorts, the specificity of DUD transmission increased substantially over time, while the genetic correlation between AUD and DUD significantly decreased. CONCLUSIONS Raised when alcohol was the preferred substance of abuse and illicit drugs highly stigmatized, AUD in parents reflected a general liability to substance use disorders, as they transmitted similar genetic risk for AUD and DUD to their children raised when both substances were widely available and relatively acceptable. DUD in parents, by contrast, reflected a more specific liability to DUD and, when transmitted to offspring, produced a considerably stronger risk for DUD than for AUD that increased over time. The magnitude and specificity of the genetic liability to psychoactive substances can be influenced by the availability of that substance.
Collapse
Affiliation(s)
- Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Linda Abrahamsson
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Henrik Ohlsson
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
Farmer RF, Seeley JR, Kosty DB, Gau JM. Deconstructing the heterogeneity of alcohol use disorder: lifetime comorbid non-alcohol substance use disorder as a distinct behavioral phenotype? Psychol Med 2023; 53:4962-4976. [PMID: 35781344 DOI: 10.1017/s0033291722001921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is an etiologically and clinically heterogeneous condition. Accumulating evidence suggests that persons with lifetime histories of comorbid AUD and non-alcohol substance use disorder (DRUG) constitute an important subgroup of AUD. This study evaluated the distinctiveness of the comorbid AUD/DRUG behavioral phenotype in a community sample with respect to risk factors, AUD course features, and outcome variables assessed at age 30. Contrast groups included persons with histories of AUD only, DRUG only, and neither AUD nor DRUG. METHODS This research utilized a prospective study design with an age-based cohort (n = 732). Participants completed four comprehensive diagnostic evaluations during the high-risk periods of adolescence, emerging adulthood, and young adulthood. RESULTS The comorbid AUD/DRUG group was distinguished from the AUD only group by risk factors, AUD course features, and outcomes. Group differences in outcomes were also explained by overall substance use disorder (SUD) severity. Persons with AUD/DRUG comorbidity were indistinguishable from those with DRUG only histories with respect to risk factors and outcomes but demonstrated greater overall SUD severity. Persons with AUD only were indistinguishable from those with neither AUD nor DRUG histories in risk factor endorsements and were mostly similar in outcomes. CONCLUSIONS Findings collectively suggest that young adults with histories of AUD only and those with comorbid AUD/DRUG are drawn from dissimilar populations. Similarities between the AUD only group with those absent AUD or DRUG histories are likely related to the former group's developmentally limited AUD course accompanied by relatively few or short-lived alcohol-related problems.
Collapse
Affiliation(s)
- Richard F Farmer
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
| | - John R Seeley
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
- College of Education, University of Oregon, 901 East 18th Ave., Eugene, OR 97403, USA
| | - Derek B Kosty
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
- College of Education, University of Oregon, 901 East 18th Ave., Eugene, OR 97403, USA
| | - Jeff M Gau
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
- College of Education, University of Oregon, 901 East 18th Ave., Eugene, OR 97403, USA
| |
Collapse
|
15
|
Vanyukov MM. Stigmata that are desired: Contradictions in addiction. ADDICTION RESEARCH & THEORY 2023; 32:83-92. [PMID: 38523739 PMCID: PMC10957146 DOI: 10.1080/16066359.2023.2238603] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/17/2023] [Indexed: 03/26/2024]
Abstract
Many experts in the etiology, assessment, and treatment of substance use/addiction view stigma and stigmatization - negatively branding addiction and substance users - as obstacles to the solution of the substance misuse problem. Discussions on this topic impact research and policy, and result in oft-repeated calls to remove the stigma from substance use and users. The goal of the article is to analyze the stigmatization concept as applied to substance use/addiction. It is widely accepted in the literature that stigmatization negatively affects substance users because addiction stigma interferes in both seeking and receiving professional care. It is argued that the societal disapproval of substance use/addiction is inappropriate because it is a mental disorder, involving biological processes. Nonetheless, neither those processes nor negative attitudes to substance use affirm the concept of stigmatization as currently applied. This concept conflates potential mistreatment and malpractice with the prosocial justified societal disapproval of a lethally dangerous behavior. Consequently, the stigmatization concept suffers from internal contradictions, is either misleading or redundant, and may do more harm than the supposed mistreatment of substance users that stigmatization connotes. On the contrary, the justified disapproval of harmful behavior may be a factor raising individual resistance to substance use. Instead of mitigating the effects of that disapproval, it may need to be capitalized on. If it is employed explicitly, conscientiously, and professionally, its internalization may be one of the resistance mechanisms needed to achieve any progress in the still elusive prevention of substance use and addiction.
Collapse
Affiliation(s)
- Michael M Vanyukov
- Departments of Pharmaceutical Sciences, Psychiatry, and Human Genetics, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
16
|
Defoe IN, Dubas JS, van Aken MAG. A cross-national study on adolescent substance use: Intentions, peer substance use, and parent-adolescent communication. JOURNAL OF RESEARCH ON ADOLESCENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR RESEARCH ON ADOLESCENCE 2023; 33:641-655. [PMID: 36717971 DOI: 10.1111/jora.12832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 10/07/2022] [Accepted: 12/24/2022] [Indexed: 05/25/2023]
Abstract
This longitudinal two-wave cross-national study investigated whether intentions, friends' substance use, and parent-adolescent substance-use specific communication predict adolescent alcohol and cannabis use 1 year later, while estimating reversed links. The temporal order between these two substances was also examined. We used multi-group cross-lagged panel modeling on data from 2 ethnically and socioeconomically diverse samples: Sint Maarten (N = 350; Mage = 14.19) and the Netherlands (N = 602; Mage = 13.50). Results showed that in the Netherlands, cannabis use predicts more subsequent problems (alcohol use, intention to use cannabis, and affiliation with cannabis-using friends). But for Sint Maarten, alcohol use predicts more subsequent problems (cannabis use, intention to use alcohol, and affiliation with alcohol-using friends). These opposing results demonstrate that caution is warranted when generalizing results across countries.
Collapse
Affiliation(s)
- Ivy N Defoe
- University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
17
|
James LM, Georgopoulos AP. Risk assessment of substance use disorders based on the human leukocyte antigen (HLA). Sci Rep 2023; 13:8545. [PMID: 37237010 DOI: 10.1038/s41598-023-35305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Substance use disorders (SUDs) are common and costly conditions that are partially attributable to genetic factors. In light of immune system influences on neural and behavioral aspects of addiction, the present study evaluated the influence of genes involved in the human immune response, human leukocyte antigen (HLA), on SUDs. We used an immunogenetic epidemiological approach to evaluate associations between the population frequencies of 127 HLA alleles and the population prevalences of six SUDs (alcohol, amphetamine, cannabis, cocaine, opioid, and "other" dependence) in 14 countries of Continental Western Europe to identify immunogenetic profiles of each SUD and evaluate their associations. The findings revealed two primary groupings of SUDs based on their immunogenetic profiles: one group comprised cannabis and cocaine, whereas the other group comprised alcohol, amphetamines, opioids, and "other" dependence. Since each individual possesses 12 HLA alleles, the population HLA-SUD scores were subsequently used to estimate individual risk for each SUD. Overall, the findings highlight similarities and differences in immunogenetic profiles of SUDs that may influence the prevalence and co-occurrence of problematic SUDs and may contribute to assessment of SUD risk of an individual on the basis of their HLA genetic makeup.
Collapse
Affiliation(s)
- Lisa M James
- The HLA Research Group, Brain Sciences Center (11B), Department of Veterans Affairs Health Care System, Minneapolis VAHCS, One Veterans Drive, Minneapolis, MN, 55417, USA.
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| | - Apostolos P Georgopoulos
- The HLA Research Group, Brain Sciences Center (11B), Department of Veterans Affairs Health Care System, Minneapolis VAHCS, One Veterans Drive, Minneapolis, MN, 55417, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| |
Collapse
|
18
|
Dash GF, Gizer IR, Martin NG, Slutske WS. Specificity in genetic and environmental risk for prescription opioid misuse and heroin use. Psychol Med 2023; 53:1-10. [PMID: 36946318 PMCID: PMC10514228 DOI: 10.1017/s003329172300034x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Many studies aggregate prescription opioid misuse (POM) and heroin use into a single phenotype, but emerging evidence suggests that their genetic and environmental influences may be partially distinct. METHODS In total, 7164 individual twins (84.12% complete pairs; 59.81% female; mean age = 30.58 years) from the Australian Twin Registry reported their lifetime misuse of prescription opioids, stimulants, and sedatives, and lifetime use of heroin, cannabis, cocaine/crack, illicit stimulants, hallucinogens, inhalants, solvents, and dissociatives via telephone interview. Independent pathway models (IPMs) and common pathway models (CPMs) partitioned the variance of drug use phenotypes into general and drug-specific genetic (a), common environmental (c), and unique environmental factors (e). RESULTS An IPM with one general a and one general e factor and a one-factor CPM provided comparable fit to the data. General factors accounted for 55% (a = 14%, e = 41%) and 79% (a = 64%, e = 15%) of the respective variation in POM and heroin use in the IPM, and 25% (a = 12%, c = 8%, e = 5%) and 80% (a = 38%, c = 27%, e = 15%) of the respective variation in POM and heroin use in the CPM. Across both models, POM emerged with substantial drug-specific genetic influence (26-39% of total phenotypic variance; 69-74% of genetic variance); heroin use did not (0% of total phenotypic variance; 0% of genetic variance in both models). Prescription sedative misuse also demonstrated significant drug-specific genetic variance. CONCLUSIONS Genetic variation in POM, but not heroin use, is predominantly drug-specific. Misuse of prescription medications that reduce experiences of subjective distress may be partially influenced by sources of genetic variation separate from illicit drug use.
Collapse
Affiliation(s)
- Genevieve F. Dash
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
| | | | - Wendy S. Slutske
- Department of Family Medicine and Community Health and Center for Tobacco Research and Intervention, University of Wisconsin, Madison, WI 53711, USA
| |
Collapse
|
19
|
The Concept of Resistance to Substance Use and a Research Approach: The Resist! Project. Twin Res Hum Genet 2023:1-9. [PMID: 36896815 PMCID: PMC10363246 DOI: 10.1017/thg.2023.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Illicit substance use is dangerous in both acute and chronic forms, frequently resulting in lethal poisoning, addiction, and other negative consequences. Similar to research in other psychiatric conditions, whose ultimate goal is to enable effective prevention and treatment, studies in substance use are focused on factors elevating the risk for the disorder. The rapid growth of the substance use problem despite the effort invested in fighting it, however, suggests the need in changing the research approach. Instead of attempting to identify risk factors, whose neutralization is often infeasible if not impossible, it may be more promising to systematically reverse the perspective to the factors enhancing the aspect of liability to disorder that shares the same dimension but is opposite to risk, that is, resistance to substance use. Resistance factors, which enable the majority of the population to remain unaffected despite the ubiquity of psychoactive substances, may be more amenable to translation. While the resistance aspect of liability is symmetric to risk, the resistance approach requires substantial changes in sampling (high-resistance rather than high-risk) and using quantitative indices of liability. This article provides an overview and a practical approach to research in resistance to substance use/addiction, currently implemented in a NIH-funded project. The project benefits from unique opportunities afforded by the data originating from two longitudinal twin studies, the Virginia Twin Study of Adolescent and Behavioral Development and the Minnesota Twin Family Study. The methodology described is also applicable to other psychiatric disorders.
Collapse
|
20
|
Galán CA, Shaw DS, O'Rourke F, Reynolds MD, Gill A, Bogen DL, Ridenour TA. Substance Use Screening and Prevention for Adolescents in Pediatric Primary Care: A Randomized Clinical Trial using the Family Check-Up. Res Child Adolesc Psychopathol 2023; 51:151-163. [PMID: 36208361 PMCID: PMC10146025 DOI: 10.1007/s10802-022-00978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/07/2022]
Abstract
This study evaluated acceptability, engagement in prevention, and efficacy of a primary care screening-and-referral-to-prevention program to reduce substance use in early adolescence. Screening tools were the Youth Risk Index and Transmissible Liability Index and prevention consisted of the Family Check-Up (FCU). Three hundred sixty-one 10- to 13-year-olds from low resource neighborhoods (85.9% African American; 52.4% female) screened "at risk" during primary care visits and were randomized to the FCU (n = 123) or usual care (n = 238). Screening was acceptable to parents and youths: nearly 95% of each rated it as important, about 90% of each were happy with or did not mind it, and only 2.4% of parents did not want their child to be screened at their next check-up. Of parents who had a chance to receive the FCU (or waitlist-control), 87.5% followed through with researchers while 93.5% who were offered FCU engaged in it. FCU efficacy primarily involved interactions such that youth with greater risk at baseline experienced larger benefits. At 12-month follow-up, FCU was associated with 11% reduced risk of initiating a new substance per substance that had been initiated before baseline; greater reductions in tolerance of deviance among those with higher tolerance of deviance at baseline; and a main effect of reduced anxiety, but no effect for conduct problems. Pediatric well-child check-up screening can identify high-risk youth before, or in the initial stages of, problematic SU; engage families in a preventive intervention; and reduce rates of substance use and related risk factors.
Collapse
Affiliation(s)
| | | | | | | | - Anne Gill
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Ty A Ridenour
- University of Pittsburgh, Pittsburgh, PA, USA. .,Research Triangle Institute, Research Triangle Park, NC, USA. .,University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
21
|
Dash GF, Gizer IR, Slutske WS. Contextualizing prescription opioid misuse and heroin use within dimensional models of drug involvement. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 5:100123. [PMID: 36591566 PMCID: PMC9802645 DOI: 10.1016/j.dadr.2022.100123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Prescription opioid misuse (POM) is often implicated in heroin initiation, despite evidence that POM does not predict heroin initiation any better than other drug use. Additionally, prescription misuse and illicit use behaviors tend to respectively "cluster" together. This study aimed to test a series of theory-driven factor models to explore how POM and heroin use are situated within the broader constellation of drug use that typically occurs alongside opioid (mis)use. Methods 36,309 individuals from NESARC-III (56.31% female; mean age=45.63 [SD=17.53]) reported their lifetime (mis)use of prescription opioids, prescription stimulants, prescription sedatives, heroin, cannabis, cocaine/crack, illicit stimulants (e.g., methamphetamine), club drugs, hallucinogens, and inhalants, and were administered a DSM-5 substance use disorder (SUD) assessment. Bifactor, correlated factors, and one-factor confirmatory factor models were fit using all drug use/SUD variables and subsequently compared. Results POM was most strongly correlated with prescription sedative misuse; heroin use was most strongly correlated with cocaine/crack use. All factor models fit the data well. Highly correlated factors and patterns of factor loadings suggested that POM and heroin use were most parsimoniously captured within a general factor alongside all other forms of drug use. This was also the case for SUD. Additional analyses testing an alternate factor structure provided further support for unidimensionality. Conclusions POM and heroin use, as well as prescription- and heroin-based SUDs, were neither separable nor distinctly associated. Future research should account for other drug use more comprehensively rather than isolating POM as a primary risk factor in heroin use and use disorder.
Collapse
Affiliation(s)
- Genevieve F. Dash
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA,Corresponding author at: 210 McAlester Hall, 320 S. 6th Street, Columbia, MO, 65211. (G.F. Dash)
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Wendy S. Slutske
- Center for Tobacco Research and Intervention and Department of Family Medicine and Community Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
22
|
Verweij KJH, Vink JM, Abdellaoui A, Gillespie NA, Derks EM, Treur JL. The genetic aetiology of cannabis use: from twin models to genome-wide association studies and beyond. Transl Psychiatry 2022; 12:489. [PMID: 36411281 PMCID: PMC9678872 DOI: 10.1038/s41398-022-02215-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022] Open
Abstract
Cannabis is among the most widely consumed psychoactive substances worldwide. Individual differences in cannabis use phenotypes can partly be explained by genetic differences. Technical and methodological advances have increased our understanding of the genetic aetiology of cannabis use. This narrative review discusses the genetic literature on cannabis use, covering twin, linkage, and candidate-gene studies, and the more recent genome-wide association studies (GWASs), as well as the interplay between genetic and environmental factors. Not only do we focus on the insights that these methods have provided on the genetic aetiology of cannabis use, but also on how they have helped to clarify the relationship between cannabis use and co-occurring traits, such as the use of other substances and mental health disorders. Twin studies have shown that cannabis use is moderately heritable, with higher heritability estimates for more severe phases of use. Linkage and candidate-gene studies have been largely unsuccessful, while GWASs so far only explain a small portion of the heritability. Dozens of genetic variants predictive of cannabis use have been identified, located in genes such as CADM2, FOXP2, and CHRNA2. Studies that applied multivariate methods (twin models, genetic correlation analysis, polygenic score analysis, genomic structural equation modelling, Mendelian randomisation) indicate that there is considerable genetic overlap between cannabis use and other traits (especially other substances and externalising disorders) and some evidence for causal relationships (most convincingly for schizophrenia). We end our review by discussing implications of these findings and suggestions for future work.
Collapse
Affiliation(s)
- Karin J. H. Verweij
- grid.7177.60000000084992262Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, The Netherlands
| | - Jacqueline M. Vink
- grid.5590.90000000122931605Behavioural Science Institute, Radboud University Nijmegen, Thomas van Aquinostraat 4, 6525 GD Nijmegen, The Netherlands
| | - Abdel Abdellaoui
- grid.7177.60000000084992262Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, The Netherlands
| | - Nathan A. Gillespie
- grid.224260.00000 0004 0458 8737Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, 800 East Leigh St, Suite 100, Richmond, VA 23219 USA
| | - Eske M. Derks
- grid.1049.c0000 0001 2294 1395Translational Neurogenomics, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006 Australia
| | - Jorien L. Treur
- grid.7177.60000000084992262Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
23
|
Deak JD, Zhou H, Galimberti M, Levey DF, Wendt FR, Sanchez-Roige S, Hatoum AS, Johnson EC, Nunez YZ, Demontis D, Børglum AD, Rajagopal VM, Jennings MV, Kember RL, Justice AC, Edenberg HJ, Agrawal A, Polimanti R, Kranzler HR, Gelernter J. Genome-wide association study in individuals of European and African ancestry and multi-trait analysis of opioid use disorder identifies 19 independent genome-wide significant risk loci. Mol Psychiatry 2022; 27:3970-3979. [PMID: 35879402 PMCID: PMC9718667 DOI: 10.1038/s41380-022-01709-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
Despite the large toll of opioid use disorder (OUD), genome-wide association studies (GWAS) of OUD to date have yielded few susceptibility loci. We performed a large-scale GWAS of OUD in individuals of European (EUR) and African (AFR) ancestry, optimizing genetic informativeness by performing MTAG (Multi-trait analysis of GWAS) with genetically correlated substance use disorders (SUDs). Meta-analysis included seven cohorts: the Million Veteran Program, Psychiatric Genomics Consortium, iPSYCH, FinnGen, Partners Biobank, BioVU, and Yale-Penn 3, resulting in a total N = 639,063 (Ncases = 20,686;Neffective = 77,026) across ancestries. OUD cases were defined as having a lifetime OUD diagnosis, and controls as anyone not known to meet OUD criteria. We estimated SNP-heritability (h2SNP) and genetic correlations (rg). Based on genetic correlation, we performed MTAG on OUD, alcohol use disorder (AUD), and cannabis use disorder (CanUD). A leave-one-out polygenic risk score (PRS) analysis was performed to compare OUD and OUD-MTAG PRS as predictors of OUD case status in Yale-Penn 3. The EUR meta-analysis identified three genome-wide significant (GWS; p ≤ 5 × 10-8) lead SNPs-one at FURIN (rs11372849; p = 9.54 × 10-10) and two OPRM1 variants (rs1799971, p = 4.92 × 10-09; rs79704991, p = 1.11 × 10-08; r2 = 0.02). Rs1799971 (p = 4.91 × 10-08) and another OPRM1 variant (rs9478500; p = 1.95 × 10-08; r2 = 0.03) were identified in the cross-ancestry meta-analysis. Estimated h2SNP was 12.75%, with strong rg with CanUD (rg = 0.82; p = 1.14 × 10-47) and AUD (rg = 0.77; p = 6.36 × 10-78). The OUD-MTAG resulted in a GWAS Nequivalent = 128,748 and 18 independent GWS loci, some mapping to genes or gene regions that have previously been associated with psychiatric or addiction phenotypes. The OUD-MTAG PRS accounted for 3.81% of OUD variance (beta = 0.61;s.e. = 0.066; p = 2.00 × 10-16) compared to 2.41% (beta = 0.45; s.e. = 0.058; p = 2.90 × 10-13) explained by the OUD PRS. The current study identified OUD variant associations at OPRM1, single variant associations with FURIN, and 18 GWS associations in the OUD-MTAG. The genetic architecture of OUD is likely influenced by both OUD-specific loci and loci shared across SUDs.
Collapse
Affiliation(s)
- Joseph D Deak
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Hang Zhou
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Marco Galimberti
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Daniel F Levey
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Frank R Wendt
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Sandra Sanchez-Roige
- University of California San Diego, La Jolla, CA, USA
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Emma C Johnson
- Washington University St. Louis Medical School, St. Louis, MO, USA
| | - Yaira Z Nunez
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Ditte Demontis
- Biomedicine, Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Anders D Børglum
- Biomedicine, Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Veera M Rajagopal
- Biomedicine, Aarhus University, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | | | - Rachel L Kember
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Amy C Justice
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | | | - Arpana Agrawal
- Washington University St. Louis Medical School, St. Louis, MO, USA
| | - Renato Polimanti
- Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Henry R Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Joel Gelernter
- Yale School of Medicine, New Haven, CT, USA.
- VA Connecticut Healthcare Center, West Haven, CT, USA.
| |
Collapse
|
24
|
Goldberg LR, Gould TJ. Genetic influences impacting nicotine use and abuse during adolescence: Insights from human and rodent studies. Brain Res Bull 2022; 187:24-38. [PMID: 35738503 DOI: 10.1016/j.brainresbull.2022.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/18/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Nicotine use continues to be a major public health concern, with an alarming recent rise in electronic cigarette consumption. Heritability estimates of nicotine use and abuse range from 40% to 80%, providing strong evidence that genetic factors impact nicotine addiction-relevant phenotypes. Although nicotine use during adolescence is a key factor in the development of addiction, it remains unclear how genetic factors impact adolescent nicotine use and abuse. This review will discuss studies investigating genetic factors impacting nicotine use during adolescence. Evidence from both rodent and human studies will be summarized and integrated when possible. Human adolescent studies have largely included candidate gene studies for genes identified in adult populations, such as genes involved in nicotine metabolism, nicotinic acetylcholine receptor signaling, dopaminergic signaling, and other neurotransmitter signaling systems. Alternatively, rodent studies have largely taken a discovery-based approach identifying strain differences in adolescent nicotine addiction-relevant behaviors. Here, we aim to answer the following three questions by integrating human and rodent findings: (1) Are there genetic variants that uniquely impact nicotine use during adolescence? (2) Are there genetic variants that impact both adolescent and adult nicotine use? and (3) Do genetic factors in adolescence significantly impact long-term consequences of adolescent nicotine use? Determining answers for these three questions will be critical for the development of preventative measures and treatments for adolescent nicotine use and addiction.
Collapse
Affiliation(s)
- Lisa R Goldberg
- Department of Biobehavioral Heatlh, Pennsylvania State University, University Park, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Heatlh, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
25
|
Chang XW, Sun Y, Muhai JN, Li YY, Chen Y, Lu L, Chang SH, Shi J. Common and distinguishing genetic factors for substance use behavior and disorder: an integrated analysis of genomic and transcriptomic studies from both human and animal studies. Addiction 2022; 117:2515-2529. [PMID: 35491750 DOI: 10.1111/add.15908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Genomic and transcriptomic findings greatly broaden the biological knowledge regarding substance use. However, systematic convergence and comparison evidence of genome-wide findings is lacking for substance use. Here, we combined all the genome-wide findings from both substance use behavior and disorder (SUBD) and identified common and distinguishing genetic factors for different SUBDs. METHODS Systemic literature search for genome-wide association (GWAS) and RNA-seq studies of alcohol/nicotine/drug use behavior (partially meets or not reported diagnostic criteria) and alcohol use behavior and disorder (AUBD), nicotine use behavior and disorder (NUBD) and drug use behavior and disorder (DUBD) was performed using PubMed and the GWAS catalog. Drug use was focused upon cannabis, opioid, cocaine and methamphetamine use. GWAS studies required case-control or case/cohort samples. RNA-seq studies were based on brain tissues. The genes which contained significant single nucleotide polymorphism (P ≤ 1 × 10-6 ) in GWAS and reported as significant in RNA-seq studies were extracted. Pathway enrichment was performed by using Metascape. Gene interaction networks were identified by using the Protein Interaction Network Analysis database. RESULTS Total SUBD-related 2910 genes were extracted from 75 GWAS studies (2 773 889 participants) and 17 RNA-seq studies. By overlapping the genes and pathways of AUBD, NUBD and DUBD, four shared genes (CACNB2, GRIN2B, PLXDC2 and PKNOX2), four shared pathways [two Gene Ontology (GO) terms of 'modulation of chemical synaptic transmission', 'regulation of trans-synaptic signaling', two Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of 'dopaminergic synapse', 'cocaine addiction'] were identified (significantly higher than random, P < 1 × 10-5 ). The top shared KEGG pathways (Benjamini-Hochberg-corrected P-value < 0.05) in the pairwise comparison of AUBD versus DUBD, NUBD versus DUBD, AUBD versus NUBD were 'Epstein-Barr virus infection', 'protein processing in endoplasmic reticulum' and 'neuroactive ligand-receptor interaction', respectively. We also identified substance-specific genetic factors: i.e. ADH1B and ALDH2 were unique for AUBD, while CHRNA3 and CHRNA4 were unique for NUBD. CONCLUSIONS This systematic review identifies the shared and unique genes and pathways for alcohol, nicotine and drug use behaviors and disorders at the genome-wide level and highlights critical biological processes for the common and distinguishing vulnerability of substance use behaviors and disorders.
Collapse
Affiliation(s)
- Xiang-Wen Chang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yan Sun
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jia-Na Muhai
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Yang-Yang Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yun Chen
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China.,Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Su-Hua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China.,Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China.,The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.,The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, China
| |
Collapse
|
26
|
UFR2709, an Antagonist of Nicotinic Acetylcholine Receptors, Delays the Acquisition and Reduces Long-Term Ethanol Intake in Alcohol-Preferring UChB Bibulous Rats. Biomedicines 2022; 10:biomedicines10071482. [PMID: 35884787 PMCID: PMC9312520 DOI: 10.3390/biomedicines10071482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
Alcoholism is a worldwide public health problem with high economic cost and which affects health and social behavior. It is estimated that alcoholism kills 3 million people globally, while in Chile it is responsible for around 9 thousand deaths per year. Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels expressed in the central nervous system, and they were suggested to modulate the ethanol mechanism involved in abuse and dependence. Previous work demonstrated a short-term treatment with UFR2709, a nAChRs antagonist, which reduced ethanol intake using a two-bottle free-choice paradigm in University of Chile bibulous (UChB) rats. Here, we present evidence of the UFR2709 efficacy in reducing the acquisition and long-term ethanol consumption. Our results show that UFR2709 (2.5 mg/kg i.p.) reduces the seek behavior and ethanol intake, even when the drug administration was stopped, and induced a reduction in the overall ethanol intake by around 55%. Using naïve UChB bibulous rats, we demonstrate that UFR2709 could delay and reduce the genetically adaptive impulse to seek and drink ethanol and prevent its excessive intake.
Collapse
|
27
|
Rabinowitz JA, Campos AI, Ong JS, García-Marín LM, Alcauter S, Mitchell BL, Grasby KL, Cuéllar-Partida G, Gillespie NA, Huhn AS, Martin NG, Thompson PM, Medland SE, Maher BS, Rentería ME. Shared Genetic Etiology between Cortical Brain Morphology and Tobacco, Alcohol, and Cannabis Use. Cereb Cortex 2022; 32:796-807. [PMID: 34379727 PMCID: PMC8841600 DOI: 10.1093/cercor/bhab243] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic variants associated with brain morphology and substance use behaviors (SUB). However, the genetic overlap between brain structure and SUB has not been well characterized. We leveraged GWAS summary data of 71 brain imaging measures and alcohol, tobacco, and cannabis use to investigate their genetic overlap using linkage disequilibrium score regression. We used genomic structural equation modeling to model a "common SUB genetic factor" and investigated its genetic overlap with brain structure. Furthermore, we estimated SUB polygenic risk scores (PRS) and examined whether they predicted brain imaging traits using the Adolescent Behavior and Cognitive Development (ABCD) study. We identified 8 significant negative genetic correlations, including between (1) alcoholic drinks per week and average cortical thickness, and (2) intracranial volume with age of smoking initiation. We observed 5 positive genetic correlations, including those between (1) insula surface area and lifetime cannabis use, and (2) the common SUB genetic factor and pericalcarine surface area. SUB PRS were associated with brain structure variation in ABCD. Our findings highlight a shared genetic etiology between cortical brain morphology and SUB and suggest that genetic variants associated with SUB may be causally related to brain structure differences.
Collapse
Affiliation(s)
- Jill A Rabinowitz
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Adrian I Campos
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jue-Sheng Ong
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Luis M García-Marín
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sarael Alcauter
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, México
| | - Brittany L Mitchell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| | - Katrina L Grasby
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Gabriel Cuéllar-Partida
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Andrew S Huhn
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Sarah E Medland
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| |
Collapse
|
28
|
Varela RB, Cararo JH, Tye SJ, Carvalho AF, Valvassori SS, Fries GR, Quevedo J. Contributions of epigenetic inheritance to the predisposition of major psychiatric disorders: theoretical framework, evidence, and implications. Neurosci Biobehav Rev 2022; 135:104579. [DOI: 10.1016/j.neubiorev.2022.104579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 02/08/2023]
|
29
|
Chang S, Sun Y, Wang F, Chang X, Zhang Y, Jia T, Sun H, Yue W, Wu P, Lu L, Shi J. Genome‐wide association meta‐analyses identify novel genetic risk loci and polygenic phenotype associations for heroin, methamphetamine and alcohol dependences. Clin Transl Med 2022; 12:e659. [PMID: 35075802 PMCID: PMC8787099 DOI: 10.1002/ctm2.659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/06/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Su‐Hua Chang
- NHC Key Laboratory of Mental Health (Peking University) National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) Chinese Academy of Medical Sciences Research Unit (No.2018RU006) Peking University Institute of Mental Health Peking University Peking University Sixth Hospital Beijing China
| | - Yan Sun
- National Institute on Drug Dependence Peking University Beijing China
- Beijing Key Laboratory on Drug Dependence Research, Peking University Beijing China
| | - Fan Wang
- Beijing HuiLongGuan Hospital, Peking University HuiLongGuan Clinical Medical School Beijing China
| | - Xiang‐Wen Chang
- National Institute on Drug Dependence Peking University Beijing China
| | - Ying‐Jian Zhang
- NHC Key Laboratory of Mental Health (Peking University) National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) Chinese Academy of Medical Sciences Research Unit (No.2018RU006) Peking University Institute of Mental Health Peking University Peking University Sixth Hospital Beijing China
| | - Tian‐Ye Jia
- Social, Genetic and Developmental Psychiatry Centre Psychology & Neuroscience King's College London De Crespigny Park Institute of Psychiatry London UK
- Institute of Science and Technology for Brain‐Inspired Intelligence, Ministry of Education‐Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence and Research and Research Institute of Intelligent Complex Systems Fudan University Shanghai China
| | - Hong‐Qiang Sun
- NHC Key Laboratory of Mental Health (Peking University) National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) Chinese Academy of Medical Sciences Research Unit (No.2018RU006) Peking University Institute of Mental Health Peking University Peking University Sixth Hospital Beijing China
| | - Wei‐Hua Yue
- NHC Key Laboratory of Mental Health (Peking University) National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) Chinese Academy of Medical Sciences Research Unit (No.2018RU006) Peking University Institute of Mental Health Peking University Peking University Sixth Hospital Beijing China
| | - Ping Wu
- National Institute on Drug Dependence Peking University Beijing China
- Beijing Key Laboratory on Drug Dependence Research, Peking University Beijing China
| | - Lin Lu
- NHC Key Laboratory of Mental Health (Peking University) National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) Chinese Academy of Medical Sciences Research Unit (No.2018RU006) Peking University Institute of Mental Health Peking University Peking University Sixth Hospital Beijing China
- National Institute on Drug Dependence Peking University Beijing China
| | - Jie Shi
- National Institute on Drug Dependence Peking University Beijing China
- Beijing Key Laboratory on Drug Dependence Research, Peking University Beijing China
- The State Key Laboratory of Natural and Biomimetic Drugs Peking University China
- The Key Laboratory for Neuroscience of the Ministry of Education and Health Peking University China
| |
Collapse
|
30
|
Fernàndez-Castillo N, Cabana-Domínguez J, Corominas R, Cormand B. Molecular genetics of cocaine use disorders in humans. Mol Psychiatry 2022; 27:624-639. [PMID: 34453125 PMCID: PMC8960411 DOI: 10.1038/s41380-021-01256-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022]
Abstract
Drug addiction, one of the major health problems worldwide, is characterized by the loss of control in drug intake, craving, and withdrawal. At the individual level, drugs of abuse produce serious consequences on health and have a negative impact on the family environment and on interpersonal and work relationships. At a wider scale, they have significant socio-economic and public health consequences and they cause delinquency and citizen insecurity. Cocaine, a psychostimulant substance, is one of the most used illicit drugs, especially in America, Western Europe, and Australia. Cocaine use disorders (CUD) are complex multifactorial conditions driven by both genetic and environmental influences. Importantly, not all people who use cocaine develop CUD, and this is due, at least in part, to biological factors that are encoded in the genome of individuals. Acute and repeated use of cocaine induces epigenetic and gene expression changes responsible for the neuronal adaptations and the remodeling of brain circuits that lead to the transition from use to abuse or dependence. The purpose of this review is to delineate such factors, which should eventually help to understand the inter-individual variability in the susceptibility to cocaine addiction. Heritability estimates for CUD are high and genetic risk factors for cocaine addiction have been investigated by candidate gene association studies (CGAS) and genome-wide association studies (GWAS), reviewed here. Also, the high comorbidity that exists between CUD and several other psychiatric disorders is well known and includes phenotypes like schizophrenia, aggression, antisocial or risk-taking behaviors. Such comorbidities are associated with a worse lifetime trajectory, and here we report shared genetic factors that may contribute to them. Gene expression changes and epigenetic modifications induced by cocaine use and chronic abuse in humans are addressed by reviewing transcriptomic studies performed on neuronal cells and on postmortem brains. We report some genes which expression is altered by cocaine that also bear genetic risk variants for the disorder. Finally, we have a glance to the pharmacogenetics of CUD treatments, still in early stages. A better understanding of the genetic underpinnings of CUD will foster the search of effective treatments and help to move forward to personalized medicine.
Collapse
Affiliation(s)
- Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain. .,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain. .,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Judit Cabana-Domínguez
- grid.5841.80000 0004 1937 0247Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain ,grid.5841.80000 0004 1937 0247Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia Spain ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia Spain
| | - Roser Corominas
- grid.5841.80000 0004 1937 0247Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain ,grid.5841.80000 0004 1937 0247Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia Spain ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia Spain
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain. .,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain. .,Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
31
|
Abstract
This brief communication responds to the article by Rajabi et al., recently published in Behavior Genetics. To test the hypothesis of cigarette smoking as a "gateway" for subsequent opium use and contrast it with the common liability model, Mendelian randomization analysis was applied to data obtained from an Iranian sample, using CHRNA3 rs1051730 as an instrumental variable. It is doubtful, however, if the assumptions of instrumental variable analysis hold in this case. The authors misstate both the gateway hypothesis and the common liability model. The article has many other deficiencies that diminish the veracity of its categorical conclusions that accept the causal interpretation of the "gateway hypothesis" and reject the common liability model, with which the data are fully consistent.
Collapse
Affiliation(s)
- Michael M Vanyukov
- Departments of Pharmaceutical Sciences, Psychiatry, and Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
32
|
Guttha N, Miao Z, Shamsuddin R. Towards the Development of a Substance Abuse Index (SEI) through Informatics. Healthcare (Basel) 2021; 9:healthcare9111596. [PMID: 34828641 PMCID: PMC8620603 DOI: 10.3390/healthcare9111596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Substance abuse or drug dependence is a prevalent phenomenon, and is on the rise in United States. Important contributing factors for the prevalence are the addictive nature of certain medicinal/prescriptive drugs, individual dispositions (biological, physiological, and psychological), and other external influences (e.g., pharmaceutical advertising campaigns). However, currently there is no comprehensive computational or machine learning framework that allows systematic studies of substance abuse and its factors with majority of the works using subjective surveys questionnaires and focusing on classification techniques. Lacking standardized methods and/or measures to prescribe medication and to study substance abuse makes it difficult to advance through collective research efforts. Thus, in this paper, we propose to test the feasibility of developing a, objective substance effect index, SEI, that can measure the tendency of an individual towards substance abuse. To that end, we combine the benefits of Electronics Medical Records (EMR) with machine learning technology by defining SEI as a function of EMR data and using logistics regression to obtain a closed form expression for SEI. We conduct various evaluations to validate the proposed model, and the results show that further work towards the development of SEI will not only provide researchers with standard computational measure for substance abuse, but may also allow them to study certain attribute interactions to gain further insights into substance abuse tendencies.
Collapse
Affiliation(s)
- Nikhila Guttha
- Department of Computer Science, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Zhuqi Miao
- Center for Health Systems Innovation, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Rittika Shamsuddin
- Department of Computer Science, Oklahoma State University, Stillwater, OK 74078, USA;
- Correspondence: ; Tel.: +1-405-744-5674
| |
Collapse
|
33
|
Abstract
Substance use disorders (SUDs) are prevalent and result in an array of negative consequences. They are influenced by genetic factors (h2 = ~50%). Recent years have brought substantial progress in our understanding of the genetic etiology of SUDs and related traits. The present review covers the current state of the field for SUD genetics, including the epidemiology and genetic epidemiology of SUDs, findings from the first-generation of SUD genome-wide association studies (GWAS), cautions about translating GWAS findings to clinical settings, and suggested prioritizations for the next wave of SUD genetics efforts. Recent advances in SUD genetics have been facilitated by the assembly of large GWAS samples, and the development of state-of-the-art methods modeling the aggregate effect of genome-wide variation. These advances have confirmed that SUDs are highly polygenic with many variants across the genome conferring risk, the vast majority of which are of small effect. Downstream analyses have enabled finer resolution of the genetic architecture of SUDs and revealed insights into their genetic relationship with other psychiatric disorders. Recent efforts have also prioritized a closer examination of GWAS findings that have suggested non-uniform genetic influences across measures of substance use (e.g. consumption) and problematic use (e.g. SUD). Additional highlights from recent SUD GWAS include the robust confirmation of loci in alcohol metabolizing genes (e.g. ADH1B and ALDH2) affecting alcohol-related traits, and loci within the CHRNA5-CHRNA3-CHRNB4 gene cluster influencing nicotine-related traits. Similar successes are expected for cannabis, opioid, and cocaine use disorders as sample sizes approach those assembled for alcohol and nicotine.
Collapse
Affiliation(s)
- Joseph D. Deak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Emma C. Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
The Association between a MAOB Variable Number Tandem Repeat Polymorphism and Cocaine and Opiate Addictions in Polyconsumers. Brain Sci 2021; 11:brainsci11101265. [PMID: 34679329 PMCID: PMC8534042 DOI: 10.3390/brainsci11101265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/02/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Genetic analysis of the association between alcohol, cocaine, and opiate addiction and variable number tandem repeat (VNTR) polymorphisms in monoamine oxidase B (MAOB) and serotonergic 5-hydroxytryptamine (serotonin) receptor 1B and 2C (HTR1B 21 and HTR2C) pathway genes was performed in a sample of 302 polyconsumers. Our genetic association analysis revealed a significant association between a 184 base pair (bp) VNTR polymorphism in the MAOB gene and addiction to cocaine and opiates. This work highlights new genetic marker associations in cocaine and opiate polyconsumer addictions. These data help to clarify and quantify the complex role of genetics in addictive disorders, as well as their future contribution to the prevention (genetic counselling), diagnosis (genetic diagnosis of vulnerability), and treatment (pharmacogenomics) of these disorders.
Collapse
|
35
|
O’Brien D, Long J, Quigley J, Lee C, McCarthy A, Kavanagh P. Association between electronic cigarette use and tobacco cigarette smoking initiation in adolescents: a systematic review and meta-analysis. BMC Public Health 2021; 21:954. [PMID: 34078351 PMCID: PMC8173887 DOI: 10.1186/s12889-021-10935-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/28/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND This systematic review of prospective longitudinal primary studies sought to determine whether electronic cigarette (e-cigarette) use by teenagers who had never smoked conventional tobacco cigarettes (tobacco cigarettes) at baseline was associated with subsequently commencing tobacco cigarette smoking. METHODS The review followed the principles of a systematic review and meta-analysis. A key word search identified peer-reviewed articles published between 1 January 2005 and 2 October 2019 from seven bibliographic databases and one search engine. Using pre-prepared inclusion/exclusion criteria two researchers independently screened abstracts, and subsequently, full text papers. Selected articles were quality assessed in duplicate. Data on study participants characteristics, exposure and outcome measures were recorded in an adapted Cochrane Data Extraction Form. Feasibility assessment was done to detect clinical heterogeneity and choose an approach to meta-analysis. Analysis comprised pairwise random effects meta-analyses, and sensitivity and subgroup analyses. RESULTS From the 6619 studies identified, 14 one-off primary studies in 21 articles were suitable for inclusion. The participants ages ranged from 13 to 19 years and comprised teenagers based in Europe and North America. Nine of the 14 one-off studies, with follow-up periods between 4 and 24 months, met the criteria for inclusion in a meta-analysis of the association between ever use of e-cigarettes and subsequent initiation of tobacco cigarette use. Based on primary study adjusted odds ratios, our meta-analysis calculated a 4.06 (95% confidence interval (CI): 3.00-5.48, I2 68%, 9 primary studies) times higher odds of commencing tobacco cigarette smoking for teenagers who had ever used e-cigarettes at baseline, though the odds ratio were marginally lower (to 3.71 times odds, 95%CI: 2.83-4. 86, I2 35%, 4 primary studies) when only the four high-quality studies were analysed. CONCLUSION The systematic review found that e-cigarette use was associated with commencement of tobacco cigarette smoking among teenagers in Europe and North America, identifying an important health-related harm. Given the availability and usage of e-cigarettes, this study provides added support for urgent response by policymakers to stop their use by teenagers to decrease direct harms in this susceptible population group, as well as to conserve achievements in diminishing tobacco cigarette initiation.
Collapse
Affiliation(s)
- Doireann O’Brien
- Health Research Board, Grattan House, 67-72 Lower Mount Street, Dublin 2, D02 H638 Ireland
| | - Jean Long
- Health Research Board, Grattan House, 67-72 Lower Mount Street, Dublin 2, D02 H638 Ireland
| | - Joan Quigley
- Health Research Board, Grattan House, 67-72 Lower Mount Street, Dublin 2, D02 H638 Ireland
| | - Caitriona Lee
- Health Research Board, Grattan House, 67-72 Lower Mount Street, Dublin 2, D02 H638 Ireland
| | - Anne McCarthy
- Health Research Board, Grattan House, 67-72 Lower Mount Street, Dublin 2, D02 H638 Ireland
| | - Paul Kavanagh
- Health Intelligence, Strategic Planning and Transformation, Health Service Executive, 4th Floor, Jervis House, Jervis Street, Dublin 1, D01 W596 Ireland
| |
Collapse
|
36
|
Krueger RF, Hobbs KA, Conway CC, Dick DM, Dretsch MN, Eaton NR, Forbes MK, Forbush KT, Keyes KM, Latzman RD, Michelini G, Patrick CJ, Sellbom M, Slade T, South S, Sunderland M, Tackett J, Waldman I, Waszczuk MA, Wright AG, Zald DH, Watson D, Kotov R. Validity and utility of Hierarchical Taxonomy of Psychopathology (HiTOP): II. Externalizing superspectrum. World Psychiatry 2021; 20:171-193. [PMID: 34002506 PMCID: PMC8129870 DOI: 10.1002/wps.20844] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The Hierarchical Taxonomy of Psychopathology (HiTOP) is an empirical effort to address limitations of traditional mental disorder diagnoses. These include arbitrary boundaries between disorder and normality, disorder co-occurrence in the modal case, heterogeneity of presentation within dis-orders, and instability of diagnosis within patients. This paper reviews the evidence on the validity and utility of the disinhibited externalizing and antagonistic externalizing spectra of HiTOP, which together constitute a broad externalizing superspectrum. These spectra are composed of elements subsumed within a variety of mental disorders described in recent DSM nosologies, including most notably substance use disorders and "Cluster B" personality disorders. The externalizing superspectrum ranges from normative levels of impulse control and self-assertion, to maladaptive disinhibition and antagonism, to extensive polysubstance involvement and personality psychopathology. A rich literature supports the validity of the externalizing superspectrum, and the disinhibited and antagonistic spectra. This evidence encompasses common genetic influences, environmental risk factors, childhood antecedents, cognitive abnormalities, neural alterations, and treatment response. The structure of these validators mirrors the structure of the phenotypic externalizing superspectrum, with some correlates more specific to disinhibited or antagonistic spectra, and others relevant to the entire externalizing superspectrum, underlining the hierarchical structure of the domain. Compared with traditional diagnostic categories, the externalizing superspectrum conceptualization shows improved utility, reliability, explanatory capacity, and clinical applicability. The externalizing superspectrum is one aspect of the general approach to psychopathology offered by HiTOP and can make diagnostic classification more useful in both research and the clinic.
Collapse
Affiliation(s)
| | - Kelsey A. Hobbs
- Department of PsychologyUniversity of MinnesotaMinneapolisMNUSA
| | | | - Danielle M. Dick
- Department of PsychologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Michael N. Dretsch
- US Army Medical Research Directorate ‐ WestWalter Reed Army Institute of Research, Joint Base Lewis‐McChordWAUSA
| | | | - Miriam K. Forbes
- Centre for Emotional Health, Department of PsychologyMacquarie UniversitySydneyNSWAustralia
| | | | | | | | - Giorgia Michelini
- Semel Institute for Neuroscience and Human BehaviorUniversity of California Los AngelesLos AngelesCAUSA
| | | | - Martin Sellbom
- Department of PsychologyUniversity of OtagoDunedinNew Zealand
| | - Tim Slade
- Matilda Centre for Research in Mental Health and Substance UseUniversity of SydneySydneyNSWAustralia
| | - Susan C. South
- Department of Psychological SciencesPurdue UniversityWest LafayetteINUSA
| | - Matthew Sunderland
- Matilda Centre for Research in Mental Health and Substance UseUniversity of SydneySydneyNSWAustralia
| | | | - Irwin Waldman
- Department of PsychologyEmory UniversityAtlantaGAUSA
| | | | | | - David H. Zald
- Department of PsychologyVanderbilt UniversityNashvilleTNUSA
| | - David Watson
- Department of PsychologyUniversity of Notre DameNotre DameINUSA
| | - Roman Kotov
- Department of PsychiatryStony Brook UniversityStony BrookNYUSA
| | | |
Collapse
|
37
|
Schuster RM, Potter K, Lamberth E, Rychik N, Hareli M, Allen S, Broos HC, Mustoe A, Gilman JM, Pachas G, Evins AE. Alcohol substitution during one month of cannabis abstinence among non-treatment seeking youth. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110205. [PMID: 33309538 PMCID: PMC7882030 DOI: 10.1016/j.pnpbp.2020.110205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Cannabis and alcohol use are correlated behaviors among youth. It is not known whether discontinuation of cannabis use is associated with changes in alcohol use. This study assessed alcohol use in youth before, during, and after 4 weeks of paid cannabis abstinence. METHODS Healthy, non-treatment seeking, cannabis users (n = 160), aged 14-25 years, 84% of whom used alcohol in the last month, were enrolled for a 4-week study with a 2-4 week follow-up. Participants were randomly assigned to 4 weeks of either biochemically-verified cannabis abstinence achieved through a contingency management framework (CB-Abst) or monitoring with no abstinence requirement (CB-Mon). Participants were assessed at baseline and approximately 4, 6, 10, 17, 24, and 31 days after enrollment. A follow-up visit with no cannabis abstinence requirement for CB-Abst was conducted after 2-4 weeks. RESULTS Sixty percent of individuals assigned to the CB-Abst condition increased in frequency and quantity of alcohol consumption during the 4-week period of incentivized cannabis abstinence. As a whole, CB-Abst increased by a mean of 0.6 drinking days and 0.2 drinks per day in the initial week of abstinence (p's < 0.006). There was no evidence for further increases in drinking frequency or quantity during the 30-day abstinence period (p's > 0.53). There was no change in drinking frequency or quantity during the 4-week monitoring or follow-up periods among CB-Mon. CONCLUSIONS On average, 4 weeks of incentivized (i.e., paid) cannabis abstinence among non-treatment seeking youth was associated with increased frequency and amount of alcohol use in week 1 that was sustained over 4 weeks and resolved with resumption of cannabis use. However, there was notable variability in individual-level response, with 60% increasing in alcohol use and 23% actually decreasing in alcohol use during cannabis abstinence. Findings suggest that increased alcohol use during cannabis abstinence among youth merits further study to determine whether this behavior occurs among treatment seeking youth and its clinical significance.
Collapse
Affiliation(s)
- Randi Melissa Schuster
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America.
| | - Kevin Potter
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - Erin Lamberth
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - Natali Rychik
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - Maya Hareli
- Department of Psychology, Loyola University Chicago, United States of America
| | - Sophia Allen
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - Hannah C Broos
- Department of Psychology, University of Miami, United States of America
| | - Audrey Mustoe
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - Jodi M Gilman
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Gladys Pachas
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America
| | - A Eden Evins
- Center for Addiction Medicine, Department of Psychiatry, MA General Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
38
|
Rai V, Kumar P. Methylenetetrahydrofolate reductase ( MTHFR) gene C677T (rs1801133) polymorphism and risk of alcohol dependence: a meta-analysis. AIMS Neurosci 2021; 8:212-225. [PMID: 33709025 PMCID: PMC7940109 DOI: 10.3934/neuroscience.2021011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 11/22/2020] [Indexed: 12/27/2022] Open
Abstract
Alcohol dependence is a complex neuropsychiatric disorder. Numerous studies investigated association between MTHFR gene C677T (rs1801133) polymorphism and alcohol dependence (AD), but the results of this association remain conflicting. Accordingly, authors conducted a meta-analysis to further investigate such an association. PubMed, Elsevier Science Direct and Springer Link databases were searched for studies on the association between the MTHFRC677T polymorphism and AD. Pooled odds ratio (OR) with 95% confidence interval (CI) was calculated using the fixed- or random-effects model. Statistical analysis was performed with the software program MetaAnayst and MIX.A total of 11 articles were identified through a search of electronic databases, up to February 28, 2020. The results of the present meta-analysis did not show any association between MTHFR C677T polymorphisms and AD risk (for T vs. C: OR = 1.04, 95% CI = 0.88-1.24; CT vs. CC: OR = 1.02, 95% CI = 0.62-1.68; for TT+CT vs. CC: OR = 1.10, 95% CI = 0.94-1.29; for TT vs. CC: OR = 1.01, 95% CI = 0.66-1.51; for TT vs. CT+CC: OR = 0.97, 95% CI = 0.66-1.40). Results of subgroup analysis showed no significant association between MTHFR C677T polymorphism with AD in Asian as well as in Caucasian population. In conclusion, C677T polymorphism is not a risk factor for alcohol dependence.
Collapse
Affiliation(s)
- Vandana Rai
- Human Molecular Genetics Laboratory, Department of Biotechnology, VBS Purvanchal University, Jaunpur-222 003, UP, India
| | - Pradeep Kumar
- Human Molecular Genetics Laboratory, Department of Biotechnology, VBS Purvanchal University, Jaunpur-222 003, UP, India
| |
Collapse
|
39
|
Thrul J, Rabinowitz JA, Reboussin BA, Maher BS, Ialongo NS. Adolescent cannabis and tobacco use are associated with opioid use in young adulthood-12-year longitudinal study in an urban cohort. Addiction 2021; 116:643-650. [PMID: 32692425 PMCID: PMC7855765 DOI: 10.1111/add.15183] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/08/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Cannabis, tobacco and alcohol use are prevalent among youth in the United States and may be risk factors for opioid use. The current study aimed at investigating associations between developmental trajectories of cannabis, tobacco and alcohol use in adolescence and opioid use in young adulthood in an urban cohort over the span of 12 years. DESIGN Cohort study of adolescents originally recruited for a randomized prevention trial with yearly assessments into young adulthood. SETTING Nine urban elementary schools in Baltimore, MD in the United States. PARTICIPANTS Participants (n = 583, 86.8% African American, 54.7% male) were originally recruited as first grade students. MEASUREMENTS Cannabis, tobacco and alcohol use were assessed annually from ages 14-18 years and opioid use from ages 19-26. Socio-demographics were assessed at age 6. Intervention status was also randomly assigned at age 6. Gender, race, free/reduced-priced lunch and intervention status were included as covariates in individual and sequential growth models. FINDINGS There were significant positive associations between the cannabis use intercept at age 14 and the opioid use intercept at age 19 (beta = 1.43; P = 0.028), the tobacco use intercept at age 14 and the opioid use intercept at age 19 (beta = 0.82; P = 0.042). Specifically, more frequent use of cannabis or tobacco at age 14 was associated with more frequent use of opioids at age 19. CONCLUSIONS Cannabis and tobacco use in early adolescence may be risk factors for opioid use in young adulthood among African Americans living in urban areas.
Collapse
Affiliation(s)
- Johannes Thrul
- Department of Mental Heath, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jill A. Rabinowitz
- Department of Mental Heath, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Beth A. Reboussin
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC,, USA
| | - Brion S. Maher
- Department of Mental Heath, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas S. Ialongo
- Department of Mental Heath, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
40
|
Abstract
Purpose of Review Substance use disorders (SUD) affect differentially women and men. Although the prevalence has been reported higher in men, those women with addictive disorders present a more vulnerable profile and are less likely to enter treatment than men. The aim of this paper is to present an overview of how sex and gender may influence epidemiology, clinical manifestations, social impact, and the neurobiological basis of these differences of women with SUD, based on human research. Recent Findings The differences in prevalence rates between genders are getting narrower; also, women tend to increase the amount of consumption more rapidly than men, showing an accelerated onset of the SUD (telescoping effect). In respect to clinical features, the most important differences are related to the risk of experience psychiatric comorbidity, the exposure to intimate partner violence, and the associated high risks in sexual and reproductive health; and those who are mothers and addicted to substances are at risk of losing the custody of children accumulating more adverse life events. Some of these differences can be based on neurobiological differences: pharmacokinetic response to substances, sensitivity to gonadal hormones, differences in neurobiological systems as glutamate, endocannabinoids, and genetic differences. Summary Specific research in women who use drugs is very scarce and treatments are not gender-sensitive oriented. For these reasons, it is important to guarantee access to the appropriate treatment of women who use drugs and a need for a gender perspective in the treatment and research of substance use disorders.
Collapse
|
41
|
Prakash J, Chatterjee K, Shankar S. Does application of complexity theory simplify concepts of psychiatry: Analogies and insights. Ind Psychiatry J 2021; 30:18-22. [PMID: 34483519 PMCID: PMC8395535 DOI: 10.4103/ipj.ipj_37_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022] Open
Abstract
Scientific curiosity has not been able to explain the cause of psychiatric illness based on primarily biological or social paradigm. Available literatures were explored to understand causality of psychiatric illness from perspective of physics. Theory of complexity and other relevant theories were extrapolated to address these questions. Mental illness appeared to be a complex interplay of reductionism and emergentism, genetic and epigenetics, stress and the vulnerability or the core and the periphery. Mental illness displayed complex interaction between biological trait and environmental state.
Collapse
Affiliation(s)
- Jyoti Prakash
- Department of Psychiatry, Armed Forces Medical College, Pune, Maharashtra, India
| | - K Chatterjee
- Department of Psychiatry, Armed Forces Medical College, Pune, Maharashtra, India
| | - S Shankar
- Consultant Rheumatologist and Clinical Immunologist, O/o DGAFMS, New Delhi, India
| |
Collapse
|
42
|
Koenig LB, Haber JR, Jacob T. Transitions in alcohol use over time: a survival analysis. BMC Psychol 2020; 8:115. [PMID: 33143748 PMCID: PMC7607670 DOI: 10.1186/s40359-020-00479-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/22/2020] [Indexed: 11/10/2022] Open
Abstract
Background The current study examined the predictors of the onset of alcohol use as well as predictors of remission and relapse, both from heavy drinking and from alcohol dependence. Similarities and differences in both clinical and psychosocial predictors across the transitions were examined.
Methods A sample of men from the Vietnam Era Twin Registry (N = 1769) completed an assessment of lifetime drinking history, which allowed age markers for starting and stopping different drinking patterns. The men also completed various assessments regarding personality, alcohol motives, and psychiatric diagnoses. Survival analyses were used to examine the predictors of the three transitions of onset, remission, and relapse for the phenotypes of heavy drinking and of alcohol dependence, censoring the individuals who had not yet experienced an event. Results As expected, predictors of onset for drinking, heavy drinking, and alcohol dependence were largely consistent and included externalizing symptomology, nicotine dependence, and cotwin history of drinking as risk factors. Predictors of remission from heavy drinking, somewhat similarly to remission from alcohol dependence, included the risk factor of externalizing disorders but also, as predicted, included more risk and protective factors in the psychosocial realm that were not predictors of onset. Contrary to our prediction, relapse to heavy drinking and alcohol dependence were predicted largely by unique psychosocial risk and protective factors including social and coping motives. Conclusion Current findings extend the findings of past research to remission and relapse in the later decades of life and have implications for treatment of alcohol use problems.
Collapse
Affiliation(s)
- Laura B Koenig
- Winona State University, Winona, MN, USA. .,Department of Social Sciences, Southwest Minnesota State University, 1501 State Street, Marshall, MN, 56258, USA.
| | - Jon Randolph Haber
- Veterans Affairs Palo Alto Health Care System, 795 Willow Road, MC 151J, Menlo Park, CA, 94025, USA
| | - Theodore Jacob
- Veterans Affairs Palo Alto Health Care System, 795 Willow Road, MC 151J, Menlo Park, CA, 94025, USA
| |
Collapse
|
43
|
Kramer J, Dick DM, King A, Ray LA, Sher KJ, Vena A, Vendruscolo LF, Acion L. Mechanisms of Alcohol Addiction: Bridging Human and Animal Studies. Alcohol Alcohol 2020; 55:603-607. [PMID: 32781467 DOI: 10.1093/alcalc/agaa068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/27/2022] Open
Abstract
AIM The purpose of this brief narrative review is to address the complexities and benefits of extending animal alcohol addiction research to the human domain, emphasizing Allostasis and Incentive Sensitization, two models that inform many pre-clinical and clinical studies. METHODS The work reviewed includes a range of approaches, including: a) animal and human studies that target the biology of craving and compulsive consumption; b) human investigations that utilize alcohol self-administration and alcohol challenge paradigms, in some cases across 10 years; c) questionnaires that document changes in the positive and negative reinforcing effects of alcohol with increasing severity of addiction; and d) genomic structural equation modeling based on data from animal and human studies. RESULTS Several general themes emerge from specific study findings. First, positive reinforcement is characteristic of early stage addiction and sometimes diminishes with increasing severity, consistent with both Allostasis and Incentive Sensitization. Second, evidence is less consistent for the predominance of negative reinforcement in later stages of addiction, a key tenant of Allostasis. Finally, there are important individual differences in motivation to drink at a given point in time as well as person-specific change patterns across time. CONCLUSIONS Key constructs of addiction, like stage and reinforcement, are by necessity operationalized differently in animal and human studies. Similarly, testing the validity of addiction models requires different strategies by the two research domains. Although such differences are challenging, they are not insurmountable, and there is much to be gained in understanding and treating addiction by combining pre-clinical and clinical approaches.
Collapse
Affiliation(s)
- John Kramer
- Department of Psychiatry, University of Iowa Carver College of Medicine, 200 Hawkins Dr, 1882JPP, Iowa City, IA 52242-1009, USA
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, 612 N. Lombardy St., Richmond, VA 23284, USA.,Department Human and Molecular Genetics, Virginia Commonwealth University, 806 West Franklin Street, Box 842018, Richmond, VA 23284, USA
| | - Andrea King
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S. Maryland Ave., Room L470, Chicago, IL 60637, USA
| | - Lara A Ray
- Department of Psychology, UCLA, 1285 Franz Hall, Los Angeles, CA 90095, USA
| | - Kenneth J Sher
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| | - Ashley Vena
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S. Maryland Ave., Room L470, Chicago, IL 60637, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Laura Acion
- Department of Psychiatry, University of Iowa Carver College of Medicine, 200 Hawkins Dr, 1882JPP, Iowa City, IA 52242-1009, USA.,Instituto de Cálculo, Universidad de Buenos Aires-CONICET, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| |
Collapse
|
44
|
Driver MN, Kuo SIC, Dick DM. Genetic feedback for psychiatric conditions: Where are we now and where are we going. Am J Med Genet B Neuropsychiatr Genet 2020; 183:423-432. [PMID: 32812348 PMCID: PMC8108123 DOI: 10.1002/ajmg.b.32815] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/12/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022]
Abstract
Genome-wide association studies are rapidly advancing our understanding of the genetic architecture of complex disorders, including many psychiatric conditions such as major depression, schizophrenia, and substance use disorders. One common goal of genome-wide association studies is to use findings for enhanced clinical prediction in the future, which can aid in identifying at-risk individuals to enable more effective prevention screening and treatment strategies. In order to achieve this goal, we first need to gain a better understanding of the issues surrounding the return of complex genetic results. In this article, we summarize the current literature on: (a) genetic literacy in the general population, (b) the public's interest in receiving genetic test results for psychiatric conditions, (c) how individuals react to and interpret their genotypic information for specific psychiatric conditions, and (d) gaps in our knowledge that will be critical to address as we move toward returning genotypic information for psychiatric conditions in both research and clinical settings. By reviewing extant studies, we aim to increase awareness of the potential benefits and consequences of returning genotypic information for psychiatric conditions.
Collapse
Affiliation(s)
- Morgan N. Driver
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Sally I-Chun Kuo
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia
| | - Danielle M. Dick
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia,Department of Psychology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
45
|
Does prenatal cocaine exposure predict adolescent substance use? Neurotoxicol Teratol 2020; 81:106906. [PMID: 32535083 DOI: 10.1016/j.ntt.2020.106906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 05/26/2020] [Accepted: 06/09/2020] [Indexed: 11/21/2022]
Abstract
Prenatal cocaine exposure (PCE) has rarely been examined as a predictor of substance use during late adolescence, and few studies have examined both the initiation of substance use and current substance use as outcomes. The present longitudinal study examined PCE, other prenatal exposures, and psychosocial risk factors for their association with substance use in mid to late adolescence. Adolescents (n = 150) followed since birth reported on their use of alcohol, cigarette, and cannabis every 6 months from age 15.0 to 17.5 using a computer-assisted self-administration version of the Youth Risk Behavior Survey. PCE did not predict substance use in a series of growth curve analyses. Several psychosocial risk factors were associated with adolescents' substance use. Having friends who use substances predicted past month cigarette and cannabis use as well as initiation of alcohol and cannabis use, while depressive symptoms predicted initiation of alcohol, cigarette, and cannabis use. The current findings suggest that more proximal psychosocial factors may play a greater role in adolescent substance use than prenatal substance exposure.
Collapse
|
46
|
Williams AR. Cannabis as a Gateway Drug for Opioid Use Disorder. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2020; 48:268-274. [PMID: 32631185 PMCID: PMC7359408 DOI: 10.1177/1073110520935338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cannabis use in some individuals can meaningfully introduce de novo risk for the initiation of opioid use and development of opioid use disorder. These risks may be particularly high during adolescence when cannabis use may disrupt critical periods of neurodevelopment. Current research studying the combination of genetic and environmental factors involved in substance use disorders is poorly understood. More research is needed, particularly to identify which adolescents are most at risk and to develop effective interventions addressing contributing factors such as trauma and psychiatric comorbidity.
Collapse
Affiliation(s)
- Arthur Robin Williams
- Arthur Robin Williams, M.D., M.B.E., is an Assistant Professor, Division on Substance Use Disorders, Columbia University Department of Psychiatry and a Research Scientist, New York State Psychiatric Institute
| |
Collapse
|
47
|
Johnson EC, Chang Y, Agrawal A. An update on the role of common genetic variation underlying substance use disorders. CURRENT GENETIC MEDICINE REPORTS 2020; 8:35-46. [PMID: 33457110 PMCID: PMC7810203 DOI: 10.1007/s40142-020-00184-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF THE REVIEW Sample size increases have resulted in novel and replicable loci for substance use disorders (SUDs). We summarize some of the latest insights into SUD genetics and discuss some next steps in addiction genetics. RECENT FINDINGS Genome-wide association studies have substantiated the role of previously known variants (e.g., rs1229984 in ADH1B for alcohol) and identified several novel loci for alcohol, tobacco, cannabis, opioid and cocaine use disorders. SUDs are genetically correlated with psychiatric outcomes, while liability to substance use is inconsistently associated with these outcomes and more closely associated with lifestyle factors. Specific variant associations appear to differ somewhat across populations, although similar genes and systems are implicated. SUMMARY The next decade of human genetic studies of addiction should focus on expanding to non-European populations, consider pleiotropy across SUD and with other psychiatric disorders, and leverage human and cross-species functional data to elucidate the biological mechanisms underlying SUDs.
Collapse
Affiliation(s)
- Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO
| | - Yoonhoo Chang
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, Saint Louis, MO
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
48
|
Datta U, Schoenrock SE, Bubier JA, Bogue MA, Jentsch JD, Logan RW, Tarantino LM, Chesler EJ. Prospects for finding the mechanisms of sex differences in addiction with human and model organism genetic analysis. GENES, BRAIN, AND BEHAVIOR 2020; 19:e12645. [PMID: 32012419 PMCID: PMC7060801 DOI: 10.1111/gbb.12645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Despite substantial evidence for sex differences in addiction epidemiology, addiction-relevant behaviors and associated neurobiological phenomena, the mechanisms and implications of these differences remain unknown. Genetic analysis in model organism is a potentially powerful and effective means of discovering the mechanisms that underlie sex differences in addiction. Human genetic studies are beginning to show precise risk variants that influence the mechanisms of addiction but typically lack sufficient power or neurobiological mechanistic access, particularly for the discovery of the mechanisms that underlie sex differences. Our thesis in this review is that genetic variation in model organisms are a promising approach that can complement these investigations to show the biological mechanisms that underlie sex differences in addiction.
Collapse
Affiliation(s)
- Udita Datta
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Sarah E. Schoenrock
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Jason A. Bubier
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Molly A. Bogue
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - James D. Jentsch
- Center for Systems Neurogenetics of Addiction, PsychologyState University of New York at BinghamtonBinghamtonNew York
| | - Ryan W. Logan
- Center for Systems Neurogenetics of Addiction, PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Lisa M. Tarantino
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Elissa J. Chesler
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| |
Collapse
|
49
|
Miech R, Keyes KM, O'Malley PM, Johnston LD. The great decline in adolescent cigarette smoking since 2000: consequences for drug use among US adolescents. Tob Control 2020; 29:638-643. [PMID: 31941823 DOI: 10.1136/tobaccocontrol-2019-055052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Adolescent cigarette smoking declined steadily and substantially from 2000 to 2018. This paper considers the potential consequences of this 'great decline' for the prevalence of other drug use among adolescents. METHODS Data are annual, cross-sectional, nationally representative Monitoring the Future surveys of more than 1.2 million US students in 12th, 10th and 8th grades from 2000 to 2018. Analyses include trends in the past 12 months' non-medical amphetamine, tranquillisers and opioid use overall, among ever and never cigarette smokers, and projected if adolescent cigarette smoking levels had remained at 2000 levels. RESULTS Within groups of ever and never cigarette smokers, the prevalence for each of the three substances has either changed little or overall increased in 2018 as compared with 2000. When the two groups were combined into one pool, the overall prevalence for each of the drugs declined by about half. The decline resulted from the growing group of never smokers, whose levels of non-medical drug use over the study period were at least four times lower than the levels of ever smokers. CONCLUSIONS The results support the 'gateway' prediction that declines in cigarette smoking among adolescents pull downward their non-medical use of amphetamines, tranquillisers and opioids. Continuing to reduce adolescent smoking through policy and programmatic prevention efforts should have further positive spillover effects on adolescent drug use.
Collapse
Affiliation(s)
- Richard Miech
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine M Keyes
- Columbia University, New York City, New York, USA.,Department of Epidemiology, Colulmbia University, New York, New York, United States
| | - Patrick M O'Malley
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Lloyd D Johnston
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Cannabis use for medical purposes has become increasingly common, including as treatment for mental health disorders such as anxiety. Unfortunately, the evidence examining its use in mental health has been slow to evolve, but is emerging. Given the widespread use of cannabis, it is important for both clinicians and those who suffer with anxiety to understand the effects of cannabis on symptoms of anxiety. In this review, we present recent, available evidence from animal models, clinical trials, and survey studies and evaluate the contribution of these studies to the current understanding of the role of cannabis in treating anxiety. RECENT FINDINGS In reviewing recent evidence, we observed significant inconsistencies across findings from preclinical studies. Large-scale surveys suggest that cannabis may be effective in reducing anxiety, however, these results stand in contrast to equivocal findings from clinical trials. SUMMARY The literature evaluating the efficacy of cannabis in anxiety disorders is in its infancy. The survey data is generally positive. Although, while some animal studies posit cannabis constituents to have anxiolytic effects, others suggest the opposite or null results. Few new clinical trials have been conducted recently, and the extant trials have significant flaws in methodology. Although anecdotal evidence from survey studies, and a small signal found in animal studies and single-dose clinical trials provide early support that cannabis may be effective for alleviating anxiety, ultimately, the current evidence is equivocal. More high-quality clinical trials must be published before sound conclusions regarding the efficacy of cannabis for treating anxiety can be drawn.
Collapse
|