1
|
Cai N, Verhulst B, Andreassen OA, Buitelaar J, Edenberg HJ, Hettema JM, Gandal M, Grotzinger A, Jonas K, Lee P, Mallard TT, Mattheisen M, Neale MC, Nurnberger JI, Peyrot WJ, Tucker-Drob EM, Smoller JW, Kendler KS. Assessment and ascertainment in psychiatric molecular genetics: challenges and opportunities for cross-disorder research. Mol Psychiatry 2025; 30:1627-1638. [PMID: 39730880 PMCID: PMC11919726 DOI: 10.1038/s41380-024-02878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/07/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Psychiatric disorders are highly comorbid, heritable, and genetically correlated [1-4]. The primary objective of cross-disorder psychiatric genetics research is to identify and characterize both the shared genetic factors that contribute to convergent disease etiologies and the unique genetic factors that distinguish between disorders [4, 5]. This information can illuminate the biological mechanisms underlying comorbid presentations of psychopathology, improve nosology and prediction of illness risk and trajectories, and aid the development of more effective and targeted interventions. In this review we discuss how estimates of comorbidity and identification of shared genetic loci between disorders can be influenced by how disorders are measured (phenotypic assessment) and the inclusion or exclusion criteria in individual genetic studies (sample ascertainment). Specifically, the depth of measurement, source of diagnosis, and time frame of disease trajectory have major implications for the clinical validity of the assessed phenotypes. Further, biases introduced in the ascertainment of both cases and controls can inflate or reduce estimates of genetic correlations. The impact of these design choices may have important implications for large meta-analyses of cohorts from diverse populations that use different forms of assessment and inclusion criteria, and subsequent cross-disorder analyses thereof. We review how assessment and ascertainment affect genetic findings in both univariate and multivariate analyses and conclude with recommendations for addressing them in future research.
Collapse
Affiliation(s)
- Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Brad Verhulst
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Ole A Andreassen
- Centre of Precision Psychiatry, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Karakter Child and Adolescent University Center, Nijmegen, The Netherlands
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John M Hettema
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael Gandal
- Departments of Psychiatry and Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Grotzinger
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Katherine Jonas
- Department of Psychiatry & Behavioral Health, Stony Brook University, Stony Brook, NY, USA
| | - Phil Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Travis T Mallard
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Manuel Mattheisen
- Department of Community Health and Epidemiology and Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital of Munich, Munich, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - John I Nurnberger
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wouter J Peyrot
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - 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.
| |
Collapse
|
2
|
Argentieri MA, Amin N, Nevado-Holgado AJ, Sproviero W, Collister JA, Keestra SM, Kuilman MM, Ginos BNR, Ghanbari M, Doherty A, Hunter DJ, Alvergne A, van Duijn CM. Integrating the environmental and genetic architectures of aging and mortality. Nat Med 2025; 31:1016-1025. [PMID: 39972219 PMCID: PMC11922759 DOI: 10.1038/s41591-024-03483-9] [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: 05/16/2023] [Accepted: 12/18/2024] [Indexed: 02/21/2025]
Abstract
Both environmental exposures and genetics are known to play important roles in shaping human aging. Here we aimed to quantify the relative contributions of environment (referred to as the exposome) and genetics to aging and premature mortality. To systematically identify environmental exposures associated with aging in the UK Biobank, we first conducted an exposome-wide analysis of all-cause mortality (n = 492,567) and then assessed the associations of these exposures with a proteomic age clock (n = 45,441), identifying 25 independent exposures associated with mortality and proteomic aging. These exposures were also associated with incident age-related multimorbidity, aging biomarkers and major disease risk factors. Compared with information on age and sex, polygenic risk scores for 22 major diseases explained less than 2 percentage points of additional mortality variation, whereas the exposome explained an additional 17 percentage points. Polygenic risk explained a greater proportion of variation (10.3-26.2%) compared with the exposome for incidence of dementias and breast, prostate and colorectal cancers, whereas the exposome explained a greater proportion of variation (5.5-49.4%) compared with polygenic risk for incidence of diseases of the lung, heart and liver. Our findings provide a comprehensive map of the contributions of environment and genetics to mortality and incidence of common age-related diseases, suggesting that the exposome shapes distinct patterns of disease and mortality risk, irrespective of polygenic disease risk.
Collapse
Affiliation(s)
- M Austin Argentieri
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA.
| | - Najaf Amin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | | | | | - Sarai M Keestra
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Midas M Kuilman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bigina N R Ginos
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aiden Doherty
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David J Hunter
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | | | | |
Collapse
|
3
|
Yu Y, Chen CZ, Celardo I, Tan BWZ, Hurcomb JD, Leal NS, Popovic R, Loh SHY, Martins LM. Enhancing mitochondrial one-carbon metabolism is neuroprotective in Alzheimer's disease models. Cell Death Dis 2024; 15:856. [PMID: 39582067 PMCID: PMC11586400 DOI: 10.1038/s41419-024-07179-3] [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: 04/18/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024]
Abstract
Alzheimer's disease (AD) is the most common form of age-related dementia. In AD, the death of neurons in the central nervous system is associated with the accumulation of toxic amyloid β peptide (Aβ) and mitochondrial dysfunction. Mitochondria are signal transducers of metabolic and biochemical information, and their impairment can compromise cellular function. Mitochondria compartmentalise several pathways, including folate-dependent one-carbon (1C) metabolism and electron transport by respiratory complexes. Mitochondrial 1C metabolism is linked to electron transport through complex I of the respiratory chain. Here, we analysed the proteomic changes in a fly model of AD by overexpressing a toxic form of Aβ (Aβ-Arc). We found that expressing Aβ-Arc caused alterations in components of both complex I and mitochondrial 1C metabolism. Genetically enhancing mitochondrial 1C metabolism through Nmdmc improved mitochondrial function and was neuroprotective in fly models of AD. We also found that exogenous supplementation with the 1C donor folinic acid improved mitochondrial health in both mammalian cells and fly models of AD. We found that genetic variations in MTHFD2L, the human orthologue of Nmdmc, were linked to AD risk. Additionally, Mendelian randomisation showed that increased folate intake decreased the risk of developing AD. Overall, our data suggest enhancement of folate-dependent 1C metabolism as a viable strategy to delay the progression and attenuate the severity of AD.
Collapse
Affiliation(s)
- Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
- Healthspan Biotics Ltd, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge, UK.
| | - Civia Z Chen
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Ivana Celardo
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Bryan Wei Zhi Tan
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - James D Hurcomb
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Nuno Santos Leal
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Rebeka Popovic
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Samantha H Y Loh
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - L Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
| |
Collapse
|
4
|
Huang Z, Zheng Z, Pang L, Fu K, Cheng J, Zhong M, Song L, Guo D, Chen Q, Li Y, Lv Y, Chen R, Sun X. The Association between Obstructive Sleep Apnea and Venous Thromboembolism: A Bidirectional Two-Sample Mendelian Randomization Study. Thromb Haemost 2024; 124:1061-1074. [PMID: 38631385 PMCID: PMC11518617 DOI: 10.1055/a-2308-2290] [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: 11/27/2023] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Despite previous observational studies linking obstructive sleep apnea (OSA) to venous thromboembolism (VTE), these findings remain controversial. This study aimed to explore the association between OSA and VTE, including pulmonary embolism (PE) and deep vein thrombosis (DVT), at a genetic level using a bidirectional two-sample Mendelian randomization (MR) analysis. METHODS Utilizing summary-level data from large-scale genome-wide association studies in European individuals, we designed a bidirectional two-sample MR analysis to comprehensively assess the genetic association between OSA and VTE. The inverse variance weighted was used as the primary method for MR analysis. In addition, MR-Egger, weighted median, and MR pleiotropy residual sum and outlier (MR-PRESSO) were used for complementary analyses. Furthermore, a series of sensitivity analyses were performed to ensure the validity and robustness of the results. RESULTS The initial and validation MR analyses indicated that genetically predicted OSA had no effects on the risk of VTE (including PE and DVT). Likewise, the reverse MR analysis did not find substantial support for a significant association between VTE (including PE and DVT) and OSA. Supplementary MR methods and sensitivity analyses provided additional confirmation of the reliability of the MR results. CONCLUSION Our bidirectional two-sample MR analysis did not find genetic evidence supporting a significant association between OSA and VTE in either direction.
Collapse
Affiliation(s)
- Zhihai Huang
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhenzhen Zheng
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lingpin Pang
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kaili Fu
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfen Cheng
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ming Zhong
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lingyue Song
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Dingyu Guo
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qiaoyun Chen
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanxi Li
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yongting Lv
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Riken Chen
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xishi Sun
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
5
|
Savage JE, Barr PB, Phung T, Lee YH, Zhang Y, McCutcheon VV, Ge T, Smoller JW, Davis LK, Meyers J, Porjesz B, Posthuma D, Mallard TT, Sanchez-Roige S. Genetic Heterogeneity Across Dimensions of Alcohol Use Behaviors. Am J Psychiatry 2024; 181:1006-1017. [PMID: 39380376 DOI: 10.1176/appi.ajp.20231055] [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: 10/10/2024]
Abstract
OBJECTIVE Increasingly large samples in genome-wide association studies (GWASs) for alcohol use behaviors (AUBs) have led to an influx of implicated genes, yet the clinical and functional understanding of these associations remains low, in part because most GWASs do not account for the complex and varied manifestations of AUBs. This study applied a multidimensional framework to investigate the latent genetic structure underlying heterogeneous dimensions of AUBs. METHODS Multimodal assessments (self-report, interview, electronic health records) were obtained from approximately 400,000 UK Biobank participants. GWAS was conducted for 18 distinct AUBs, including consumption, drinking patterns, alcohol problems, and clinical sequelae. Latent genetic factors were identified and carried forward to GWAS using genomic structural equation modeling, followed by functional annotation, genetic correlation, and enrichment analyses to interpret the genetic associations. RESULTS Four latent factors were identified: Problems, Consumption, BeerPref (declining alcohol consumption with a preference for drinking beer), and AtypicalPref (drinking fortified wine and spirits). The latent factors were moderately correlated (rg values, 0.12-0.57) and had distinct patterns of associations, with BeerPref in particular implicating many novel genomic regions. Patterns of regional and cell type-specific gene expression in the brain also differed between the latent factors. CONCLUSIONS Deep phenotyping is an important next step to improve understanding of the genetic etiology of AUBs, in addition to increasing sample size. Further effort is required to uncover the genetic heterogeneity underlying AUBs using methods that account for their complex, multidimensional nature.
Collapse
Affiliation(s)
- Jeanne E Savage
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Peter B Barr
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Tanya Phung
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Younga H Lee
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Yingzhe Zhang
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Vivia V McCutcheon
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Tian Ge
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Jordan W Smoller
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Lea K Davis
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Jacquelyn Meyers
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Bernice Porjesz
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Travis T Mallard
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| | - Sandra Sanchez-Roige
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam (Savage, Phung, Posthuma); Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York (Barr, Meyers, Porjesz); VA New York Harbor Healthcare System, Brooklyn, New York (Barr, Meyers); Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine (Lee, Zhang, Ge, Smoller, Mallard), and Center for Precision Psychiatry (Ge, Smoller), Massachusetts General Hospital, Boston; Department of Psychiatry, Harvard Medical School, Boston (Lee, Ge, Smoller, Mallard); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge (Lee, Zhang, Ge, Smoller, Mallard); Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston (Zhang); Department of Psychiatry, Washington University School of Medicine, St. Louis (McCutcheon); Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville (Davis, Sanchez-Roige); Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, VU University Medical Center, Amsterdam (Posthuma); Department of Psychiatry and Institute for Genomic Medicine, University of California San Diego, La Jolla (Sanchez-Roige)
| |
Collapse
|
6
|
Gorman BR, Ji SG, Francis M, Sendamarai AK, Shi Y, Devineni P, Saxena U, Partan E, DeVito AK, Byun J, Han Y, Xiao X, Sin DD, Timens W, Moser J, Muralidhar S, Ramoni R, Hung RJ, McKay JD, Bossé Y, Sun R, Amos CI, Pyarajan S. Multi-ancestry GWAS meta-analyses of lung cancer reveal susceptibility loci and elucidate smoking-independent genetic risk. Nat Commun 2024; 15:8629. [PMID: 39366959 PMCID: PMC11452618 DOI: 10.1038/s41467-024-52129-4] [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: 04/08/2024] [Accepted: 08/27/2024] [Indexed: 10/06/2024] Open
Abstract
Lung cancer remains the leading cause of cancer mortality, despite declining smoking rates. Previous lung cancer GWAS have identified numerous loci, but separating the genetic risks of lung cancer and smoking behavioral susceptibility remains challenging. Here, we perform multi-ancestry GWAS meta-analyses of lung cancer using the Million Veteran Program cohort (approximately 95% male cases) and a previous study of European-ancestry individuals, jointly comprising 42,102 cases and 181,270 controls, followed by replication in an independent cohort of 19,404 cases and 17,378 controls. We then carry out conditional meta-analyses on cigarettes per day and identify two novel, replicated loci, including the 19p13.11 pleiotropic cancer locus in squamous cell lung carcinoma. Overall, we report twelve novel risk loci for overall lung cancer, lung adenocarcinoma, and squamous cell lung carcinoma, nine of which are externally replicated. Finally, we perform PheWAS on polygenic risk scores for lung cancer, with and without conditioning on smoking. The unconditioned lung cancer polygenic risk score is associated with smoking status in controls, illustrating a reduced predictive utility in non-smokers. Additionally, our polygenic risk score demonstrates smoking-independent pleiotropy of lung cancer risk across neoplasms and metabolic traits.
Collapse
Affiliation(s)
- Bryan R Gorman
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Sun-Gou Ji
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- BridgeBio Pharma, Palo Alto, CA, USA
| | - Michael Francis
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Anoop K Sendamarai
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Yunling Shi
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Poornima Devineni
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Uma Saxena
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth Partan
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Andrea K DeVito
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- University Medical Centre Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, Netherlands
- Department of Pathology & Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Jennifer Moser
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Sumitra Muralidhar
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Rachel Ramoni
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - James D McKay
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Ryan Sun
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Savage JE, de Leeuw CA, Werme J, Dick DM, Posthuma D, van der Sluis S. Refining the scope of genetic influences on alcohol misuse through environmental stratification and gene-environment interaction. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1853-1865. [PMID: 39198719 PMCID: PMC11661684 DOI: 10.1111/acer.15425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Gene-environment interaction (G × E) is likely an important influence shaping individual differences in alcohol misuse (AM), yet it has not been extensively studied in molecular genetic research. In this study, we use a series of genome-wide gene-environment interaction (GWEIS) and in silico annotation methods with the aim of improving gene identification and biological understanding of AM. METHODS We carried out GWEIS for four AM phenotypes in the large UK Biobank sample (N = 360,314), with trauma exposure and socioeconomic status (SES) as moderators of the genetic effects. Exploratory analyses compared stratified genome-wide association (GWAS) and GWEIS modeling approaches. We applied functional annotation, gene- and gene-set enrichment, and polygenic score analyses to interpret the GWEIS results. RESULTS GWEIS models showed few genetic variants with significant interaction effects across gene-environment pairs. Enrichment analyses identified moderation by SES of the genes NOXA1, DLGAP1, and UBE2L3 on drinking quantity and the gene IFIT1B on drinking frequency. Except for DLGAP1, these genes have not previously been linked to AM. The most robust results (GWEIS interaction p = 4.59e-09) were seen for SES moderating the effects of variants linked to immune-related genes on a pattern of drinking with versus without meals. CONCLUSIONS Our results highlight several genes and a potential mechanism of immune system functioning behind the moderating effect of SES on the genetic influences on AM. Although GWEIS seems to be a preferred approach over stratified GWAS, modeling G × E effects at the molecular level remains a challenge even in large samples. Understanding these effects will require substantial effort and more in-depth phenotypic measurement.
Collapse
Affiliation(s)
- Jeanne E. Savage
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Christiaan A. de Leeuw
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Josefin Werme
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | | | - Danielle M. Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers Addiction Research Center, Rutgers University, Piscataway, NJ, USA
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Sophie van der Sluis
- Department of Child and Adolescent Psychology and Psychiatry, section Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Naito T, Inoue K, Namba S, Sonehara K, Suzuki K, Matsuda K, Kondo N, Toda T, Yamauchi T, Kadowaki T, Okada Y. Machine learning reveals heterogeneous associations between environmental factors and cardiometabolic diseases across polygenic risk scores. COMMUNICATIONS MEDICINE 2024; 4:181. [PMID: 39304733 PMCID: PMC11415376 DOI: 10.1038/s43856-024-00596-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Although polygenic risk scores (PRSs) are expected to be helpful in precision medicine, it remains unclear whether high-PRS groups are more likely to benefit from preventive interventions for diseases. Recent methodological advancements enable us to predict treatment effects at the individual level. METHODS We employed causal forest to explore the relationship between PRSs and individual risk of diseases associated with certain environmental factors. Following simulations illustrating its performance, we applied our approach to investigate the individual risk of cardiometabolic diseases, including coronary artery diseases (CAD) and type 2 diabetes (T2D), associated with obesity and smoking among individuals from UK Biobank (UKB; n = 369,942) and BioBank Japan (BBJ; n = 149,421). RESULTS Here we find the heterogeneous association of obesity and smoking with diseases across PRS values, complicated by the multi-dimensional combination of individual characteristics such as age and sex. The highest positive correlations of PRSs and the exposure-related disease risks are observed between obesity and T2D in UKB and between smoking and CAD in BBJ (Spearman's ρ = 0.61 and 0.32, respectively). However, most relationships are weak or negative, suggesting that high-PRS groups will not necessarily benefit most from environmental factor prevention. CONCLUSIONS Our study highlights the importance of individual-level prediction of disease risks associated with target exposure in precision medicine.
Collapse
Affiliation(s)
- Tatsuhiko Naito
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan.
| | - Kosuke Inoue
- Department of Social Epidemiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Hakubi Center, Kyoto University, Kyoto, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken Suzuki
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoki Kondo
- Department of Social Epidemiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan.
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan.
| |
Collapse
|
9
|
Jones AG, Connelly GG, Dalapati T, Wang L, Schott BH, San Roman AK, Ko DC. Biological sex affects functional variation across the human genome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.03.24313025. [PMID: 39281750 PMCID: PMC11398442 DOI: 10.1101/2024.09.03.24313025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Humans display sexual dimorphism across many traits, but little is known about underlying genetic mechanisms and impacts on disease. We utilized single-cell RNA-seq of 480 lymphoblastoid cell lines to reveal that the vast majority (79%) of sex-biased genes are targets of transcription factors that display sex-biased expression. Further, we developed a two-step regression method that identified sex-biased expression quantitative trait loci (sb-eQTL) across the genome. In contrast to previous work, these sb-eQTL are abundant (n=10,754; FDR 5%) and reproducible (replication up to π1=0.56). These sb-eQTL are enriched in over 600 GWAS phenotypes, including 120 sb-eQTL associated with the female-biased autoimmune disease multiple sclerosis. Our results demonstrate widespread genetic impacts on sexual dimorphism and identify possible mechanisms and clinical targets for sex differences in diverse diseases.
Collapse
Affiliation(s)
- Angela G. Jones
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
- Duke University Program in Genetics and Genomics, Duke University; Durham, NC, USA
| | - Guinevere G. Connelly
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
- Duke University Program in Genetics and Genomics, Duke University; Durham, NC, USA
| | - Trisha Dalapati
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
| | - Benjamin H. Schott
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
- Duke University Program in Genetics and Genomics, Duke University; Durham, NC, USA
| | - Adrianna K. San Roman
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
- Duke University Program in Genetics and Genomics, Duke University; Durham, NC, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University; Durham, NC, USA
- Duke University Program in Genetics and Genomics, Duke University; Durham, NC, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University; Durham, NC, USA
| |
Collapse
|
10
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zalesky A, Davatzikos C. The genetic architecture of biological age in nine human organ systems. NATURE AGING 2024; 4:1290-1307. [PMID: 38942983 PMCID: PMC11446180 DOI: 10.1038/s43587-024-00662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/30/2024] [Indexed: 06/30/2024]
Abstract
Investigating the genetic underpinnings of human aging is essential for unraveling the etiology of and developing actionable therapies for chronic diseases. Here, we characterize the genetic architecture of the biological age gap (BAG; the difference between machine learning-predicted age and chronological age) across nine human organ systems in 377,028 participants of European ancestry from the UK Biobank. The BAGs were computed using cross-validated support vector machines, incorporating imaging, physical traits and physiological measures. We identify 393 genomic loci-BAG pairs (P < 5 × 10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary and renal systems. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system (organ specificity) while exerting pleiotropic links with other organ systems (interorgan cross-talk). We find that genetic correlation between the nine BAGs mirrors their phenotypic correlation. Further, a multiorgan causal network established from two-sample Mendelian randomization and latent causal variance models revealed potential causality between chronic diseases (for example, Alzheimer's disease and diabetes), modifiable lifestyle factors (for example, sleep duration and body weight) and multiple BAGs. Our results illustrate the potential for improving human organ health via a multiorgan network, including lifestyle interventions and drug repurposing strategies.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Xue A, Zhu Z, Wang H, Jiang L, Visscher PM, Zeng J, Yang J. Unravelling the complex causal effects of substance use behaviours on common diseases. COMMUNICATIONS MEDICINE 2024; 4:43. [PMID: 38472333 DOI: 10.1038/s43856-024-00473-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Substance use behaviours (SUB) including smoking, alcohol consumption, and coffee intake are associated with many health outcomes. However, whether the health effects of SUB are causal remains controversial, especially for alcohol consumption and coffee intake. METHODS In this study, we assess 11 commonly used Mendelian Randomization (MR) methods by simulation and apply them to investigate the causal relationship between 7 SUB traits and health outcomes. We also combine stratified regression, genetic correlation, and MR analyses to investigate the dosage-dependent effects. RESULTS We show that smoking initiation has widespread risk effects on common diseases such as asthma, type 2 diabetes, and peripheral vascular disease. Alcohol consumption shows risk effects specifically on cardiovascular diseases, dyslipidemia, and hypertensive diseases. We find evidence of dosage-dependent effects of coffee and tea intake on common diseases (e.g., cardiovascular disease and osteoarthritis). We observe that the minor allele effect of rs4410790 (the top signal for tea intake level) is negative on heavy tea intake ( b ̂ G W A S = - 0.091 , s . e . = 0.007 , P = 4.90 × 10 - 35 ) but positive on moderate tea intake ( b ̂ G W A S = 0.034 , s . e . = 0.006 , P = 3.40 × 10 - 8 ) , compared to the non-tea-drinkers. CONCLUSION Our study reveals the complexity of the health effects of SUB and informs design for future studies aiming to dissect the causal relationships between behavioural traits and complex diseases.
Collapse
Affiliation(s)
- Angli Xue
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zhihong Zhu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- National Centre for Register-Based Research, Aarhus University, Aarhus V, 8210, Denmark
| | - Huanwei Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Longda Jiang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jian Zeng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
| |
Collapse
|
12
|
Clive J, Flintham E, Savolainen V. Same-sex sociosexual behaviour is widespread and heritable in male rhesus macaques. Nat Ecol Evol 2023; 7:1287-1301. [PMID: 37429903 DOI: 10.1038/s41559-023-02111-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/01/2023] [Indexed: 07/12/2023]
Abstract
Numerous reports have documented the occurrence of same-sex sociosexual behaviour (SSB) across animal species. However, the distribution of the behaviour within a species needs to be studied to test hypotheses describing its evolution and maintenance, in particular whether the behaviour is heritable and can therefore evolve by natural selection. Here we collected detailed observations across 3 yr of social and mounting behaviour of 236 male semi-wild rhesus macaques, which we combined with a pedigree dating back to 1938, to show that SSB is both repeatable (19.35%) and heritable (6.4%). Demographic factors (age and group structure) explained SSB variation only marginally. Furthermore, we found a positive genetic correlation between same-sex mounter and mountee activities, indicating a common basis to different forms of SSB. Finally, we found no evidence of fitness costs to SSB, but show instead that the behaviour mediated coalitionary partnerships that have been linked to improved reproductive success. Together, our results demonstrate that SSB is frequent in rhesus macaques, can evolve, and is not costly, indicating that SSB may be a common feature of primate reproductive ecology.
Collapse
Affiliation(s)
- Jackson Clive
- Georgina Mace Centre for the Living Planet, Department of Life Sciences, Silwood Park Campus, Imperial College London, Ascot, UK
| | - Ewan Flintham
- Georgina Mace Centre for the Living Planet, Department of Life Sciences, Silwood Park Campus, Imperial College London, Ascot, UK
| | - Vincent Savolainen
- Georgina Mace Centre for the Living Planet, Department of Life Sciences, Silwood Park Campus, Imperial College London, Ascot, UK.
| |
Collapse
|
13
|
White JD, Bierut LJ. Alcohol Consumption and Alcohol Use Disorder: Exposing an Increasingly Shared Genetic Architecture. Am J Psychiatry 2023; 180:530-532. [PMID: 37525606 PMCID: PMC10765608 DOI: 10.1176/appi.ajp.20230456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Affiliation(s)
- Julie D White
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, N.C. (White); Department of Psychiatry, Washington University School of Medicine, St. Louis (Bierut)
| | - Laura J Bierut
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, N.C. (White); Department of Psychiatry, Washington University School of Medicine, St. Louis (Bierut)
| |
Collapse
|
14
|
D'Antona S, Pathak GA, Koller D, Porro D, Cava C, Polimanti R. Phenome-wide genetic-correlation analysis and genetically informed causal inference of amyotrophic lateral sclerosis. Hum Genet 2023; 142:1173-1183. [PMID: 36773064 PMCID: PMC10449723 DOI: 10.1007/s00439-023-02525-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/18/2023] [Indexed: 02/12/2023]
Abstract
Leveraging genome-wide association statistics generated from a large study of amyotrophic lateral sclerosis (ALS; 29,612 cases and 122,656 controls) and UK Biobank (UKB; 4,024 phenotypes, up to 361,194 participants), we conducted a phenome-wide analysis of ALS genetic liability and identified 46 genetically correlated traits, such as fluid intelligence score (rg = - 0.21, p = 1.74 × 10-6), "spending time in pub or social club" (rg = 0.24, p = 2.77 × 10-6), non-work related walking (rg = - 0.25, p = 1.95 × 10-6), college education (rg = - 0.15, p = 7.08 × 10-5), "ever diagnosed with panic attacks (rg = 0.39, p = 4.24 × 10-5), and "self-reported other gastritis including duodenitis" (rg = 0.28, p = 1.4 × 10-3). To assess the putative directionality of these genetic correlations, we conducted a latent causal variable analysis, identifying significant genetic causality proportions (gĉp) linking ALS genetic liability to seven traits. While the genetic component of "self-reported other gastritis including duodenitis" showed a causal effect on ALS (gĉp = 0.50, p = 1.26 × 10-29), the genetic liability to ALS is potentially causal for multiple traits, also including an effect on "ever being diagnosed with panic attacks" (gĉp = 0.79, p = 5.011 × 10-15) and inverse effects on "other leisure/social group activities" (gĉp = 0.66, p = 1 × 10-4) and prospective memory result (gĉp = 0.35, p = 0.005). Our subsequent Mendelian randomization analysis indicated that some of these associations may be due to bidirectional effects. In conclusion, this phenome-wide investigation of ALS polygenic architecture highlights the widespread pleiotropy linking this disorder with several health domains.
Collapse
Affiliation(s)
- Salvatore D'Antona
- Institute of Bioimaging and Molecular Physiology, National Research Council, Milan, Italy
| | - Gita A Pathak
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Dora Koller
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Catalonia, Spain
| | - Danilo Porro
- Institute of Bioimaging and Molecular Physiology, National Research Council, Milan, Italy
| | - Claudia Cava
- Institute of Bioimaging and Molecular Physiology, National Research Council, Milan, Italy.
| | - Renato Polimanti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
15
|
Mignogna G, Carey CE, Wedow R, Baya N, Cordioli M, Pirastu N, Bellocco R, Malerbi KF, Nivard MG, Neale BM, Walters RK, Ganna A. Patterns of item nonresponse behaviour to survey questionnaires are systematic and associated with genetic loci. Nat Hum Behav 2023; 7:1371-1387. [PMID: 37386106 PMCID: PMC10444625 DOI: 10.1038/s41562-023-01632-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/17/2023] [Indexed: 07/01/2023]
Abstract
Response to survey questionnaires is vital for social and behavioural research, and most analyses assume full and accurate response by participants. However, nonresponse is common and impedes proper interpretation and generalizability of results. We examined item nonresponse behaviour across 109 questionnaire items in the UK Biobank (N = 360,628). Phenotypic factor scores for two participant-selected nonresponse answers, 'Prefer not to answer' (PNA) and 'I don't know' (IDK), each predicted participant nonresponse in follow-up surveys (incremental pseudo-R2 = 0.056), even when controlling for education and self-reported health (incremental pseudo-R2 = 0.046). After performing genome-wide association studies of our factors, PNA and IDK were highly genetically correlated with one another (rg = 0.73 (s.e. = 0.03)) and with education (rg,PNA = -0.51 (s.e. = 0.03); rg,IDK = -0.38 (s.e. = 0.02)), health (rg,PNA = 0.51 (s.e. = 0.03); rg,IDK = 0.49 (s.e. = 0.02)) and income (rg,PNA = -0.57 (s.e. = 0.04); rg,IDK = -0.46 (s.e. = 0.02)), with additional unique genetic associations observed for both PNA and IDK (P < 5 × 10-8). We discuss how these associations may bias studies of traits correlated with item nonresponse and demonstrate how this bias may substantially affect genome-wide association studies. While the UK Biobank data are deidentified, we further protected participant privacy by avoiding exploring non-response behaviour to single questions, assuring that no information can be used to associate results with any particular respondents.
Collapse
Affiliation(s)
- Gianmarco Mignogna
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Caitlin E Carey
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Robbee Wedow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Sociology, Purdue University, West Lafayette, IN, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- AnalytiXIN (Analytics Indiana), Indianapolis, IN, USA.
- Department of Statistics, Purdue University, West Lafayette, IN, USA.
| | - Nikolas Baya
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mattia Cordioli
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Fondazione Human Technopole, Viale Rita Levi-Montalcini, Milan, Italy
| | - Rino Bellocco
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Michel G Nivard
- Department of Biological Psychiatry, Faculty of Behavioural and Movement Sciences, Vrije Universiteit, Amsterdam, the Netherlands
- Methodology Program, Amsterdam Public Health, Amsterdam, the Netherlands
- Amsterdam Neuroscience - Mood, Anxiety, Psychosis, Stress and Sleep, Amsterdam, the Netherlands
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrea Ganna
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
Liang S, Wang N, Wang Y, Wang M, Zhao X, Yang M, Yi H, Zhu M, Wang C, Hang D, Jiang Y, Dai J. Polygenic risk for termination of the 'healthspan' and its interactions with lifestyle factors: A prospective cohort study based on 288,359 participants. Maturitas 2023; 175:107786. [PMID: 37354644 DOI: 10.1016/j.maturitas.2023.107786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
OBJECTIVES To investigate whether a polygenic risk score (PRS) and its interactions with lifestyle factors are associated with termination of the 'healthspan' (the number of years living without serious diseases or degeneration). DESIGN, EXPOSURES AND PARTICIPANTS Death or the incidence of any of seven independent morbidities (cancer, congestive heart failure, myocardial infarction, chronic obstructive pulmonary disease, stroke, dementia, and diabetes) strongly associated with aging were considered to define the termination of the healthspan. A total of 288,359 healthy participants from the UK Biobank were included in this prospective cohort study to evaluate the associations between PRS, lifestyle, and healthspan. The PRS was generated by weighting 12 healthspan-related genetic loci, which and scores were then categorized into three groups in Cox regression models. A lifestyle index was developed that incorporated body mass index (BMI), alcohol consumption, diet, smoking, and physical activity, and these scores were also categorized into three groups. The risk of termination of the healthspan was calculated across the different PRS and lifestyle index groups using Cox regression models. Interactions were estimated with the marginal effect of lifestyle on the risk of termination of healthspan across values of the moderator PRS using kernel estimation. RESULTS During an average follow-up of 9.83 years, 68,903 healthspan-termination events occurred. It was calculated that people with high polygenic risk could reduce their risk of healthspan termination by 40 % if they maintain a favorable lifestyle. The marginal effect of lifestyle on the risk of healthspan termination increased with growing genetic risk. Smoking and diet showed monotonic changes in opposite directions, while BMI, physical activity, and alcohol had a U-shaped interaction with genetic risk. CONCLUSIONS Favorable lifestyle can attenuate the risk of termination of the healthspan, especially for people with high genetic risk. The improvement afforded by ideal lifestyle behaviors varies for each individual.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Nanxi Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoyu Zhao
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meiqi Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Honggang Yi
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, China
| | - Dong Hang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
17
|
Flint J. The genetic basis of major depressive disorder. Mol Psychiatry 2023; 28:2254-2265. [PMID: 36702864 PMCID: PMC10611584 DOI: 10.1038/s41380-023-01957-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023]
Abstract
The genetic dissection of major depressive disorder (MDD) ranks as one of the success stories of psychiatric genetics, with genome-wide association studies (GWAS) identifying 178 genetic risk loci and proposing more than 200 candidate genes. However, the GWAS results derive from the analysis of cohorts in which most cases are diagnosed by minimal phenotyping, a method that has low specificity. I review data indicating that there is a large genetic component unique to MDD that remains inaccessible to minimal phenotyping strategies and that the majority of genetic risk loci identified with minimal phenotyping approaches are unlikely to be MDD risk loci. I show that inventive uses of biobank data, novel imputation methods, combined with more interviewer diagnosed cases, can identify loci that contribute to the episodic severe shifts of mood, and neurovegetative and cognitive changes that are central to MDD. Furthermore, new theories about the nature and causes of MDD, drawing upon advances in neuroscience and psychology, can provide handles on how best to interpret and exploit genetic mapping results.
Collapse
Affiliation(s)
- Jonathan Flint
- Department of Psychiatry and Biobehavioral Sciences, Billy and Audrey Wilder Endowed Chair in Psychiatry and Neuroscience, Center for Neurobehavioral Genetics, 695 Charles E. Young Drive South, 3357B Gonda, Box 951761, Los Angeles, CA, 90095-1761, USA.
| |
Collapse
|
18
|
Chen S, Yang F, Xu T, Wang Y, Zhang K, Fu G, Zhang W. Smoking and coronary artery disease risk in patients with diabetes: A Mendelian randomization study. Front Immunol 2023; 14:891947. [PMID: 36776880 PMCID: PMC9910331 DOI: 10.3389/fimmu.2023.891947] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Background Previous observational studies have shown an association between smoking and coronary artery disease (CAD) in patients with diabetes. Whether this association reflects causality remains unestablished. This study aimed to explore the causal effect of smoking on CAD in patients with diabetes. Methods Genetic signatures for smoking were extracted from a large genome-wide association study (GWAS), consisted of up to 1.2 million participants. Four smoking phenotypes were included: smoking initiation, cigarettes per day, age at initiation of regular smoking, and smoking cessation. Genetic associations with CAD in patients with diabetes were extracted from another GWAS, which included 15,666 participants (3,968 CAD cases and 11,696 controls). The analyses were performed using the univariable and multivariable Mendelian randomization (MR) method. Results MR analysis revealed that smoking initiation was positively related to CAD risk in patients with diabetes (OR = 1.322, 95% CI = 1.114 - 1.568, P = 0.001), but this association was attenuated when adjusted for cardiovascular risk factors (OR = 1.212, 95% CI = 1.008 - 1.457, P = 0.041). Age at initiation of regular smoking was negatively related to CAD in patients with diabetes (OR = 0.214, 95% CI = 0.070 - 0.656, P = 0.007), but this association became insignificant when adjusted for cardiovascular risk factors. Conclusions This study supported the effect of smoking initiation on the risk of CAD in patients with diabetes.
Collapse
Affiliation(s)
- Songzan Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Fangkun Yang
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Tian Xu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yao Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Kaijie Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China,*Correspondence: Wenbin Zhang,
| |
Collapse
|
19
|
Guo H, Yu X, Liu Y, Paik DT, Justesen JM, Chandy M, Jahng JWS, Zhang T, Wu W, Rwere F, Zhao SR, Pokhrel S, Shivnaraine RV, Mukherjee S, Simon DJ, Manhas A, Zhang A, Chen CH, Rivas MA, Gross ER, Mochly-Rosen D, Wu JC. SGLT2 inhibitor ameliorates endothelial dysfunction associated with the common ALDH2 alcohol flushing variant. Sci Transl Med 2023; 15:eabp9952. [PMID: 36696485 DOI: 10.1126/scitranslmed.abp9952] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The common aldehyde dehydrogenase 2 (ALDH2) alcohol flushing variant known as ALDH2*2 affects ∼8% of the world's population. Even in heterozygous carriers, this missense variant leads to a severe loss of ALDH2 enzymatic activity and has been linked to an increased risk of coronary artery disease (CAD). Endothelial cell (EC) dysfunction plays a determining role in all stages of CAD pathogenesis, including early-onset CAD. However, the contribution of ALDH2*2 to EC dysfunction and its relation to CAD are not fully understood. In a large genome-wide association study (GWAS) from Biobank Japan, ALDH2*2 was found to be one of the strongest single-nucleotide polymorphisms associated with CAD. Clinical assessment of endothelial function showed that human participants carrying ALDH2*2 exhibited impaired vasodilation after light alcohol drinking. Using human induced pluripotent stem cell-derived ECs (iPSC-ECs) and CRISPR-Cas9-corrected ALDH2*2 iPSC-ECs, we modeled ALDH2*2-induced EC dysfunction in vitro, demonstrating an increase in oxidative stress and inflammatory markers and a decrease in nitric oxide (NO) production and tube formation capacity, which was further exacerbated by ethanol exposure. We subsequently found that sodium-glucose cotransporter 2 inhibitors (SGLT2i) such as empagliflozin mitigated ALDH2*2-associated EC dysfunction. Studies in ALDH2*2 knock-in mice further demonstrated that empagliflozin attenuated ALDH2*2-mediated vascular dysfunction in vivo. Mechanistically, empagliflozin inhibited Na+/H+-exchanger 1 (NHE-1) and activated AKT kinase and endothelial NO synthase (eNOS) pathways to ameliorate ALDH2*2-induced EC dysfunction. Together, our results suggest that ALDH2*2 induces EC dysfunction and that SGLT2i may potentially be used as a preventative measure against CAD for ALDH2*2 carriers.
Collapse
Affiliation(s)
- Hongchao Guo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuan Yu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johanne Marie Justesen
- Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James W S Jahng
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tiejun Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weijun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Freeborn Rwere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suman Pokhrel
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | - Daniel J Simon
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amit Manhas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manuel A Rivas
- Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Eric R Gross
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
20
|
Deak JD, Levey DF, Wendt FR, Zhou H, Galimberti M, Kranzler HR, Gaziano JM, Stein MB, Polimanti R, Gelernter J. Genome-Wide Investigation of Maximum Habitual Alcohol Intake in US Veterans in Relation to Alcohol Consumption Traits and Alcohol Use Disorder. JAMA Netw Open 2022; 5:e2238880. [PMID: 36301540 PMCID: PMC9614582 DOI: 10.1001/jamanetworkopen.2022.38880] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/30/2022] [Indexed: 11/14/2022] Open
Abstract
Importance Alcohol genome-wide association studies (GWASs) have generally focused on alcohol consumption and alcohol use disorder (AUD); few have examined habitual drinking behaviors like maximum habitual alcohol intake (MaxAlc). Objectives To identify genetic loci associated with MaxAlc and to elucidate the genetic architecture across alcohol traits. Design, Setting, and Participants This MaxAlc genetic association study was performed among Million Veteran Program participants enrolled from January 10, 2011, to September 30, 2020. Ancestry-specific GWASs were conducted in participants with European (n = 218 623) and African (n = 29 132) ancestry, then meta-analyzed (N = 247 755). Linkage-disequilibrium score regression was used to estimate single nucleotide variant (SNV)-heritability and genetic correlations (rg) with other alcohol and psychiatric traits. Genomic structural equation modeling (gSEM) was used to evaluate genetic associations between MaxAlc and other alcohol traits. Mendelian randomization was used to examine potential causal relationships between MaxAlc and liver enzyme levels. MTAG (multitrait analysis of GWAS) was used to analyze MaxAlc and problematic alcohol use (PAU) jointly. Exposures Genetic associations. Main Outcomes and Measures MaxAlc was defined from the following survey item: "in a typical month, what is/was the largest number of drinks of alcohol you may have had in one day?" with ordinal responses from 0 to 15 or more drinks. Results GWASs were conducted on sample sizes of as many as 247 455 US veterans. Participants were 92.68% male and had mean (SD) age of 65.92 (11.70) years. The MaxAlc GWAS resulted in 15 genome-wide significant loci. Top associations in European-ancestry and African-ancestry participants were with known functional variants in the ADH1B gene, namely rs1229984 (P = 3.12 × 10-101) and rs2066702 (P = 6.30 × 10-17), respectively. Novel associations were also found. SNV-heritability was 6.65% (SE, 0.41) in European-ancestry participants and 3.42% (SE, 1.46) in African-ancestry participants. MaxAlc was positively correlated with PAU (rg = 0.79; P = 3.95 × 10-149) and AUD (rg = 0.76; P = 1.26 × 10-127) and had negative rg with the UK Biobank "alcohol usually taken with meals" (rg = -0.53; P = 1.40 × 10-50). For psychiatric traits, MaxAlc had the strongest genetic correlation with suicide attempt (rg = 0.40; P = 3.02 × 10-21). gSEM supported a 2-factor model with MaxAlc loading on a factor with PAU and AUD and other alcohol consumption measures loading on a separate factor. Mendelian randomization supported an association between MaxAlc and the liver enzyme gamma-glutamyltransferase (β = 0.012; P = 2.66 × 10-10). MaxAlc MTAG resulted in 31 genome-wide significant loci. Conclusions and Relevance The findings suggest that MaxAlc closely aligns genetically with PAU traits. This study improves understanding of the mechanisms associated with normative alcohol consumption vs problematic habitual use and AUD as well as how MaxAlc relates to psychiatric and medical conditions genetically and biologically.
Collapse
Affiliation(s)
- Joseph D. Deak
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Daniel F. Levey
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Frank R. Wendt
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Hang Zhou
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Marco Galimberti
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Henry R. Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia
- Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - J. Michael Gaziano
- Massachusetts Veterans Epidemiology and Research Information Center (MAVERIC), Boston Veterans Affairs Healthcare System, Boston
- Department of Medicine, Divisions of Aging and Preventative Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Murray B. Stein
- University of California, San Diego, La Jolla
- VA San Diego Healthcare System, San Diego, California
| | - Renato Polimanti
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| | - Joel Gelernter
- Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare Center, West Haven, Connecticut
| |
Collapse
|
21
|
Bell JA, Richardson TG, Wang Q, Sanderson E, Palmer T, Walker V, O'Keeffe LM, Timpson NJ, Cichonska A, Julkunen H, Würtz P, Holmes MV, Davey Smith G. Effects of general and central adiposity on circulating lipoprotein, lipid, and metabolite levels in UK Biobank: A multivariable Mendelian randomization study. THE LANCET REGIONAL HEALTH. EUROPE 2022; 21:100457. [PMID: 35832062 PMCID: PMC9272390 DOI: 10.1016/j.lanepe.2022.100457] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Background The direct effects of general adiposity (body mass index (BMI)) and central adiposity (waist-to-hip-ratio (WHR)) on circulating lipoproteins, lipids, and metabolites are unknown. Methods We used new metabolic data from UK Biobank (N=109,532, a five-fold higher N over previous studies). EDTA-plasma was used to quantify 249 traits with nuclear-magnetic-resonance spectroscopy including subclass-specific lipoprotein concentrations and lipid content, plus pre-glycemic and inflammatory metabolites. We used univariable and multivariable two-stage least-squares regression models with genetic risk scores for BMI and WHR as instruments to estimate total (unadjusted) and direct (mutually-adjusted) effects of BMI and WHR on metabolic traits; plus effects on statin use and interaction by sex, statin use, and age (proxy for medication use). Findings Higher BMI decreased apolipoprotein B and low-density lipoprotein cholesterol (LDL-C) before and after WHR-adjustment, whilst BMI increased triglycerides only before WHR-adjustment. These effects of WHR were larger and BMI-independent. Direct effects differed markedly by sex, e.g., triglycerides increased only with BMI among men, and only with WHR among women. Adiposity measures increased statin use and showed metabolic effects which differed by statin use and age. Among the youngest (38-53y, statins-5%), BMI and WHR (per-SD) increased LDL-C (total effects: 0.04-SD, 95%CI=-0.01,0.08 and 0.10-SD, 95%CI=0.02,0.17 respectively), but only WHR directly. Among the oldest (63-73y, statins-29%), BMI and WHR directly lowered LDL-C (-0.19-SD, 95%CI=-0.27,-0.11 and -0.05-SD, 95%CI=-0.16,0.06 respectively). Interpretation Excess adiposity likely raises atherogenic lipid and metabolite levels exclusively via adiposity stored centrally, particularly among women. Apparent effects of adiposity on lowering LDL-C are likely explained by an effect of adiposity on statin use. Funding UK Medical Research Council; British Heart Foundation; Novo Nordisk; National Institute for Health Research; Wellcome Trust; Cancer Research UK.
Collapse
Affiliation(s)
- Joshua A. Bell
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom G. Richardson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Novo Nordisk Research Centre Oxford, Old Road Campus, Oxford, UK
| | - Qin Wang
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom Palmer
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Venexia Walker
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Linda M. O'Keeffe
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Public Health, Western Gateway Building, University College Cork, Ireland
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | | | - Michael V. Holmes
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit at the University of Oxford, Oxford, UK
- National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
22
|
Sanchez-Roige S, Kember RL, Agrawal A. Substance use and common contributors to morbidity: A genetics perspective. EBioMedicine 2022; 83:104212. [PMID: 35970022 PMCID: PMC9399262 DOI: 10.1016/j.ebiom.2022.104212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Excessive substance use and substance use disorders (SUDs) are common, serious and relapsing medical conditions. They frequently co-occur with other diseases that are leading contributors to disability worldwide. While heavy substance use may potentiate the course of some of these illnesses, there is accumulating evidence suggesting common genetic architectures. In this narrative review, we focus on four heritable medical conditions - cardiometabolic disease, chronic pain, depression and COVID-19, which are commonly overlapping with, but not necessarily a direct consequence of, SUDs. We find persuasive evidence of underlying genetic liability that predisposes to both SUDs and chronic pain, depression, and COVID-19. For cardiometabolic disease, there is greater support for a potential causal influence of problematic substance use. Our review encourages de-stigmatization of SUDs and the assessment of substance use in clinical settings. We assert that identifying shared pathways of risk has high translational potential, allowing tailoring of treatments for multiple medical conditions. FUNDING: SSR acknowledges T29KT0526, T32IR5226 and DP1DA054394; RLK acknowledges AA028292; AA acknowledges DA054869 & K02DA032573. The funders had no role in the conceptualization or writing of the paper.
Collapse
Affiliation(s)
- Sandra Sanchez-Roige
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Rachel L Kember
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
23
|
Pirastu N, McDonnell C, Grzeszkowiak EJ, Mounier N, Imamura F, Merino J, Day FR, Zheng J, Taba N, Concas MP, Repetto L, Kentistou KA, Robino A, Esko T, Joshi PK, Fischer K, Ong KK, Gaunt TR, Kutalik Z, Perry JRB, Wilson JF. Using genetic variation to disentangle the complex relationship between food intake and health outcomes. PLoS Genet 2022; 18:e1010162. [PMID: 35653391 PMCID: PMC9162356 DOI: 10.1371/journal.pgen.1010162] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 03/22/2022] [Indexed: 02/02/2023] Open
Abstract
Diet is considered as one of the most important modifiable factors influencing human health, but efforts to identify foods or dietary patterns associated with health outcomes often suffer from biases, confounding, and reverse causation. Applying Mendelian randomization in this context may provide evidence to strengthen causality in nutrition research. To this end, we first identified 283 genetic markers associated with dietary intake in 445,779 UK Biobank participants. We then converted these associations into direct genetic effects on food exposures by adjusting them for effects mediated via other traits. The SNPs which did not show evidence of mediation were then used for MR, assessing the association between genetically predicted food choices and other risk factors, health outcomes. We show that using all associated SNPs without omitting those which show evidence of mediation, leads to biases in downstream analyses (genetic correlations, causal inference), similar to those present in observational studies. However, MR analyses using SNPs which have only a direct effect on the exposure on food exposures provided unequivocal evidence of causal associations between specific eating patterns and obesity, blood lipid status, and several other risk factors and health outcomes.
Collapse
Affiliation(s)
- Nicola Pirastu
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Human Technopole, Milan, Italy
| | - Ciara McDonnell
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Cardiovascular Sciences, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Eryk J. Grzeszkowiak
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Ninon Mounier
- Centre for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Fumiaki Imamura
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Jordi Merino
- Diabetes Unit and Centre for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Felix R. Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, United Kingdom
| | - Nele Taba
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste, Italy
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Katherine A. Kentistou
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Cardiovascular Sciences, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Peter K. Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Krista Fischer
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ken K. Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Tom R. Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, United Kingdom
| | - Zoltán Kutalik
- Centre for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - John R. B. Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - James F. Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
24
|
Bountress KE, Brick LA, Sheerin C, Grotzinger A, Bustamante D, Hawn SE, Gillespie N, Kirkpatrick RM, Kranzler H, Morey R, Edenberg HJ, Maihofer AX, Disner S, Ashley-Koch A, Peterson R, Lori A, Stein DJ, Kimbrel N, Nievergelt C, Andreassen OA, Luykx J, Javanbakht A, Youssef NA, Amstadter AB. Alcohol use and alcohol use disorder differ in their genetic relationships with PTSD: A genomic structural equation modelling approach. Drug Alcohol Depend 2022; 234:109430. [PMID: 35367939 PMCID: PMC9018560 DOI: 10.1016/j.drugalcdep.2022.109430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/20/2022]
Abstract
PURPOSE Posttraumatic Stress Disorder (PTSD) is associated with increased alcohol use and alcohol use disorder (AUD), which are all moderately heritable. Studies suggest the genetic association between PTSD and alcohol use differs from that of PTSD and AUD, but further analysis is needed. BASIC PROCEDURES We used genomic Structural Equation Modeling (genomicSEM) to analyze summary statistics from large-scale genome-wide association studies (GWAS) of European Ancestry participants to investigate the genetic relationships between PTSD (both diagnosis and re-experiencing symptom severity) and a range of alcohol use and AUD phenotypes. MAIN FINDINGS When we differentiated genetic factors for alcohol use and AUD we observed improved model fit relative to models with all alcohol-related indicators loading onto a single factor. The genetic correlations (rG) of PTSD were quite discrepant for the alcohol use and AUD factors. This was true when modeled as a three-correlated-factor model (PTSD-AUD rG:.36, p < .001; PTSD-alcohol use rG: -0.17, p < .001) and as a Bifactor model, in which the common and unique portions of alcohol phenotypes were pulled out into an AUD-specific factor (rG with PTSD:.40, p < .001), AU-specific factor (rG with PTSD: -0.57, p < .001), and a common alcohol factor (rG with PTSD:.16, NS). PRINCIPAL CONCLUSIONS These results indicate the genetic architecture of alcohol use and AUD are differentially associated with PTSD. When the portions of variance unique to alcohol use and AUD are extracted, their genetic associations with PTSD vary substantially, suggesting different genetic architectures of alcohol phenotypes in people with PTSD.
Collapse
Affiliation(s)
- Kaitlin E Bountress
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA.
| | - Leslie A Brick
- Department of Psychiatry and Human Behavior, Quantitative Sciences Program, Alpert Medical School at Brown University, USA
| | - Christina Sheerin
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA
| | - Andrew Grotzinger
- Behavioral, Psychiatric, and Statistical Genetics, Institute for Behavior Genetics, University of Colorado Boulder, USA
| | - Daniel Bustamante
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA; Integrative Life Sciences Doctoral Program, Virginia Commonwealth University, USA
| | - Sage E Hawn
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Nathan Gillespie
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA
| | - Robert M Kirkpatrick
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA
| | - Henry Kranzler
- University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, PA, USA; Mental Illness Research, Education and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Rajendra Morey
- VA Mid-Atlantic Mental Illness Research Education and Clinical Center, Durham VAMC, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke-UNC Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Adam X Maihofer
- Department of Psychiatry, University of California, San Diego, USA; Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Seth Disner
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Allison Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Roseann Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University, USA
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Nathan Kimbrel
- VA Mid-Atlantic Mental Illness Research Education and Clinical Center, Durham VAMC, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Caroline Nievergelt
- Department of Psychiatry, University of California, San Diego, USA; Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Jurjen Luykx
- School for Mental Health and Neuroscience, Maastricht University Medical Centre, Department of Psychiatry and Neuropsychology Maastricht, The Netherlands; UMC Utrecht Brain Center, University Medical Center Utrecht, Department of Psychiatry Utrecht, University, Utrecht, The Netherlands; Outpatient second opinion clinic, GGNet Mental Health, Warnsveld, The Netherlands
| | - Arash Javanbakht
- Stress, Trauma, and Anxiety Research Clinic (STARC), Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, USA
| | - Nagy A Youssef
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Ananda B Amstadter
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, USA
| |
Collapse
|
25
|
Nagpal S, Tandon R, Gibson G. Canalization of the Polygenic Risk for Common Diseases and Traits in the UK Biobank Cohort. Mol Biol Evol 2022; 39:6547257. [PMID: 35275999 PMCID: PMC9004416 DOI: 10.1093/molbev/msac053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Since organisms develop and thrive in the face of constant perturbations due to environmental and genetic variation, species may evolve resilient genetic architectures. We sought evidence for this process, known as canalization, through a comparison of the prevalence of phenotypes as a function of the polygenic score (PGS) across environments in the UK Biobank cohort study. Contrasting seven diseases and three categorical phenotypes with respect to 151 exposures in 408,925 people, the deviation between the prevalence-risk curves was observed to increase monotonically with the PGS percentile in one-fifth of the comparisons, suggesting extensive PGS-by-Environment (PGS×E) interaction. After adjustment for the dependency of allelic effect sizes on increased prevalence in the perturbing environment, cases where polygenic influences are greater or lesser than expected are seen to be particularly pervasive for educational attainment, obesity, and metabolic condition type-2 diabetes. Inflammatory bowel disease analysis shows fewer interactions but confirms that smoking and some aspects of diet influence risk. Notably, body mass index has more evidence for decanalization (increased genetic influence at the extremes of polygenic risk), whereas the waist-to-hip ratio shows canalization, reflecting different evolutionary pressures on the architectures of these weight-related traits. An additional 10 % of comparisons showed evidence for an additive shift of prevalence independent of PGS between exposures. These results provide the first widespread evidence for canalization protecting against disease in humans and have implications for personalized medicine as well as understanding the evolution of complex traits. The findings can be explored through an R shiny app at https://canalization-gibsonlab.shinyapps.io/rshiny/.
Collapse
Affiliation(s)
- Sini Nagpal
- School of Biological Sciences, and Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Raghav Tandon
- Wallace H. Coulter Department of Biomedical Engineering, and Center for Machine Learning, Georgia Institute of Technology, Atlanta, GA, USA
| | - Greg Gibson
- School of Biological Sciences, and Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
26
|
Wang X, Jia M, Mao Y, Jia Z, Liu H, Yang G, Wang S, Sun B, Zhang H. Very-light alcohol consumption suppresses breast tumor progression in a mouse model. Food Funct 2022; 13:3391-3404. [PMID: 35230367 DOI: 10.1039/d1fo02089g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The relationship between alcohol consumption and cancer has no consistent results both in epidemiological studies and animal models. The inaccuracy of alcohol consumption dosage in the experimental design maybe leads to inconsistent results and makes the researchers ignore the effect of very-light alcohol consumption on cancer. To determine the effects of very-light alcohol consumption on cancer, in this study, the manner of gavage was used to control the alcohol consumption accurately. The impacts of age and time of drinking on cancer progression were also evaluated in this study. Here, we find that a certain range of alcohol consumption (from 0.5% w/v to 2.0% w/v) can suppress tumor development in the breast metastasis mouse model by controlling the alcohol consumption dosage accurately. RNA sequencing analyses were performed in primary tumors and related metastases from the NC group and 1.0% w/v group. The results of primary tumors and related metastases indicated that chronic very-light alcohol consumption downregulates breast tumor-associated oncogenes in primary tumors and regulates the immune system and metabolic system in metastatic carcinoma. To provide the public with drinking recommendations, eight commercial alcohol types were investigated at a dosage of 1.0% w/v. Two types of commercial alcohol, red wine (made in France, brand 1) and baijiu (made in China, brand 1), exerted excellent primary tumor and metastasis inhibitory effects. The untargeted metabolomic analysis of commercial alcohol by liquid chromatography-tandem mass spectrometry indicated that baijiu (brand 1) and baijiu (brand 2) exhibited a difference in compositions that can lead to their different anti-cancer effects. These results indicated that a certain range of very light alcohol dosages might have a potential human-cancer inhibition effect.
Collapse
Affiliation(s)
- Xiuxiu Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Min Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Yifei Mao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Huilin Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China.
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Shuo Wang
- School of Medicine, Nankai University, Tianjin 300457, China.
| | - Baoguo Sun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China.
| | - Hongyan Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China.
| |
Collapse
|
27
|
Jamshidi J, Schofield PR, Gatt JM, Fullerton JM. Phenotypic and genetic analysis of a wellbeing factor score in the UK Biobank and the impact of childhood maltreatment and psychiatric illness. Transl Psychiatry 2022; 12:113. [PMID: 35304435 PMCID: PMC8933416 DOI: 10.1038/s41398-022-01874-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 01/07/2023] Open
Abstract
Wellbeing is an important aspect of mental health that is moderately heritable. Specific wellbeing-related variants have been identified via GWAS meta-analysis of individual questionnaire items. However, a multi-item within-subject index score has potential to capture greater heritability, enabling improved delineation of genetic and phenotypic relationships across traits and exposures that are not possible on aggregate-data. This research employed data from the UK Biobank resource, and a wellbeing index score was derived from indices of happiness and satisfaction with family/friendship/finances/health, using principal component analysis. GWAS was performed in Caucasian participants (N = 129,237) using the derived wellbeing index, followed by polygenic profiling (independent sample; N = 23,703). The wellbeing index, its subcomponents, and negative indicators of mental health were compared via phenotypic and genetic correlations, and relationships with psychiatric disorders examined. Lastly, the impact of childhood maltreatment on wellbeing was investigated. Five independent genome-wide significant loci for wellbeing were identified. The wellbeing index had SNP-heritability of ~8.6%, and stronger phenotypic and genetic correlations with its subcomponents (0.55-0.77) than mental health phenotypes (-0.21 to -0.39). The wellbeing score was lower in participants reporting various psychiatric disorders compared to the total sample. Childhood maltreatment exposure was also associated with reduced wellbeing, and a moderate genetic correlation (rg = ~-0.56) suggests an overlap in heritability of maltreatment with wellbeing. Thus, wellbeing is negatively associated with both psychiatric disorders and childhood maltreatment. Although notable limitations, biases and assumptions are discussed, this within-cohort study aids the delineation of relationships between a quantitative wellbeing index and indices of mental health and early maltreatment.
Collapse
Affiliation(s)
- Javad Jamshidi
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Justine M Gatt
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia. .,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
28
|
Hop PJ, Zwamborn RA, Hannon E, Shireby GL, Nabais MF, Walker EM, van Rheenen W, van Vugt JJ, Dekker AM, Westeneng HJ, Tazelaar GH, van Eijk KR, Moisse M, Baird D, Khleifat AA, Iacoangeli A, Ticozzi N, Ratti A, Cooper-Knock J, Morrison KE, Shaw PJ, Basak AN, Chiò A, Calvo A, Moglia C, Canosa A, Brunetti M, Grassano M, Gotkine M, Lerner Y, Zabari M, Vourc’h P, Corcia P, Couratier P, Pardina JSM, Salas T, Dion P, Ross JP, Henderson RD, Mathers S, McCombe PA, Needham M, Nicholson G, Rowe DB, Pamphlett R, Mather KA, Sachdev PS, Furlong S, Garton FC, Henders AK, Lin T, Ngo ST, Steyn FJ, Wallace L, Williams KL, Neto MM, Cauchi RJ, Blair IP, Kiernan MC, Drory V, Povedano M, de Carvalho M, Pinto S, Weber M, Rouleau GA, Silani V, Landers JE, Shaw CE, Andersen PM, McRae AF, van Es MA, Pasterkamp RJ, Wray NR, McLaughlin RL, Hardiman O, Kenna KP, Tsai E, Runz H, Al-Chalabi A, van den Berg LH, Van Damme P, Mill J, Veldink JH. Genome-wide study of DNA methylation shows alterations in metabolic, inflammatory, and cholesterol pathways in ALS. Sci Transl Med 2022; 14:eabj0264. [PMID: 35196023 PMCID: PMC10040186 DOI: 10.1126/scitranslmed.abj0264] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with an estimated heritability between 40 and 50%. DNA methylation patterns can serve as proxies of (past) exposures and disease progression, as well as providing a potential mechanism that mediates genetic or environmental risk. Here, we present a blood-based epigenome-wide association study meta-analysis in 9706 samples passing stringent quality control (6763 patients, 2943 controls). We identified a total of 45 differentially methylated positions (DMPs) annotated to 42 genes, which are enriched for pathways and traits related to metabolism, cholesterol biosynthesis, and immunity. We then tested 39 DNA methylation-based proxies of putative ALS risk factors and found that high-density lipoprotein cholesterol, body mass index, white blood cell proportions, and alcohol intake were independently associated with ALS. Integration of these results with our latest genome-wide association study showed that cholesterol biosynthesis was potentially causally related to ALS. Last, DNA methylation at several DMPs and blood cell proportion estimates derived from DNA methylation data were associated with survival rate in patients, suggesting that they might represent indicators of underlying disease processes potentially amenable to therapeutic interventions.
Collapse
Affiliation(s)
- Paul J. Hop
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Ramona A.J. Zwamborn
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Eilis Hannon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Gemma L. Shireby
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Marta F. Nabais
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Emma M. Walker
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Joke J.F.A. van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Annelot M. Dekker
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Gijs H.P. Tazelaar
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Kristel R. van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Matthieu Moisse
- KU Leuven–University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven 3000, Belgium
- VIB, Center for Brain and Disease Research, Leuven 3000, Belgium
- University Hospitals Leuven, Department of Neurology, Leuven 3000, Belgium
| | - Denis Baird
- Translational Biology, Biogen, Boston, MA 02142, USA
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King’s College London, London SE5 8AZ, UK
| | - Nicola Ticozzi
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan 20122, Italy
| | - Antonia Ratti
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano 20145, Italy
| | - Jonathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - Karen E. Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - A. Nazli Basak
- Koc University, School of Medicine, Translational Medicine Research Center, NDAL, Istanbul, 34450, Turkey
| | - Adriano Chiò
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Andrea Calvo
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Cristina Moglia
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Antonio Canosa
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Maura Brunetti
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
| | - Maurizio Grassano
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
| | - Marc Gotkine
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Yossef Lerner
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Michal Zabari
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Patrick Vourc’h
- Service de Biochimie et Biologie moléculaire, CHU de Tours, Tours 37044, France
- UMR 1253, Université de Tours, Inserm, Tours 37044, France
| | - Philippe Corcia
- UMR 1253, Université de Tours, Inserm, Tours 37044, France
- Centre de référence sur la SLA, CHU de Tours, Tours 37044, France
| | - Philippe Couratier
- Centre de référence sur la SLA, CHRU de Limoges, Limoges 87042, France
- UMR 1094, Université de Limoges, Inserm, Limoges 87025, France
| | | | - Teresa Salas
- Department of Neurology, Hospital La Paz-Carlos III, Madrid 28046, Spain
| | - Patrick Dion
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
| | - Jay P. Ross
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3A 0C7, Canada
| | - Robert D. Henderson
- Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
| | - Susan Mathers
- Calvary Health Care Bethlehem, Parkdale, VIC 3195, Australia
| | - Pamela A. McCombe
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
| | - Merrilee Needham
- Fiona Stanley Hospital, Perth, WA 6150, Australia
- Notre Dame University, Fremantle, WA 6160, Australia
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA 6150, Australia
| | - Garth Nicholson
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Dominic B. Rowe
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Roger Pamphlett
- Discipline of Pathology and Department of Neuropathology, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Karen A. Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2031, Australia
- Neuroscience Research Australia Institute, Randwick, NSW 2031, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2031, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, UNSW, Randwick, NSW 2031, Australia
| | - Sarah Furlong
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Fleur C. Garton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Anjali K. Henders
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Tian Lin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Shyuan T. Ngo
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Frederik J. Steyn
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Kelly L. Williams
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | | | | | | | - Ruben J. Cauchi
- Center for Molecular Medicine and Biobanking and Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta
| | - Ian P. Blair
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Matthew C. Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, 2050, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Vivian Drory
- Department of Neurology, Tel-Aviv Sourasky Medical Centre, Tel-Aviv 64239, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Monica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Mamede de Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Susana Pinto
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Markus Weber
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Guy A. Rouleau
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan 20122, Italy
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Christopher E. Shaw
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Peter M. Andersen
- Department of Clinical Science, Umeå University, Umeå SE-901 85, Sweden
| | - Allan F. McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Michael A. van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, 3584 CX, Netherlands
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Russell L. McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Kevin P. Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, 3584 CX, Netherlands
| | - Ellen Tsai
- Translational Biology, Biogen, Boston, MA 02142, USA
| | - Heiko Runz
- Translational Biology, Biogen, Boston, MA 02142, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- King’s College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Leonard H. van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Philip Van Damme
- KU Leuven–University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven 3000, Belgium
- VIB, Center for Brain and Disease Research, Leuven 3000, Belgium
- University Hospitals Leuven, Department of Neurology, Leuven 3000, Belgium
| | - Jonathan Mill
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Jan H. Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| |
Collapse
|
29
|
Cheng X, Ouyang F, Ma T, Luo Y, Yin J, Li J, Zhang G, Bai Y. Association of Healthy Lifestyle and Life Expectancy in Patients With Cardiometabolic Multimorbidity: A Prospective Cohort Study of UK Biobank. Front Cardiovasc Med 2022; 9:830319. [PMID: 35757322 PMCID: PMC9218816 DOI: 10.3389/fcvm.2022.830319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background The prevalence of cardiometabolic multimorbidity (CMM), which significantly increases the risk of mortality, is increasing globally. However, the role of healthy lifestyle in the secondary prevention of CMM is unclear. Methods In total, 290,795 participants with CMM, which was defined as coexistence of at least two of hypertension (HTN), diabetes mellitus (DM), coronary heart disease (CHD), and stroke (ST), and those without these four diseases at baseline were derived from UK Biobank. The associations between specific CMM patterns and mortality, and that between healthy lifestyle (including physical activity, smoking, alcohol consumption, and vegetable and fruit consumption) and mortality in patients with specific CMM patterns were calculated using the flexible parametric Royston-Parmar proportion-hazard model. Hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were calculated. Results During a median 12.3-year follow up period, 15,537 (5.3%) deaths occurred. Compared with participants without cardiometabolic diseases, the HRs for all-cause mortality were 1.54 [95% confidence interval (CI): 1.30, 1.82] in participants with HTN + DM, 1.84 (95% CI: 1.59, 2.12) in those with HTN + CHD, 1.89 (95% CI: 1.46, 2.45) in those with HTN + ST, and 2.89 (95% CI: 2.28, 3.67) in those with HTN + DM + CHD. At the age of 45 years, non-current smoking was associated with an increase in life expectancy by 3.72, 6.95, 6.75, and 4.86 years for participants with HTN + DM, HTN + CHD, HTN + ST, and HTN + DM + CHD, respectively. A corresponding increase by 2.03, 1.95, 2.99, and 1.88 years, respectively, was observed in participants with regular physical activity. Non-/moderate alcohol consumption and adequate fruit/vegetable consumption were not significantly associated with life expectancy in patients with specific CMM patterns. Conclusion Cardiometabolic multimorbidity was associated with an increased risk of mortality. Regular physical activity and non-current smoking can increase life expectancy in patients with specific CMM patterns.
Collapse
Affiliation(s)
- Xunjie Cheng
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Feiyun Ouyang
- Department of Social Medicine and Health Management, Central South University, Changsha, China
| | - Tianqi Ma
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Luo
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China
| | - Jinghua Yin
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China
| | - Jinchen Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongping Bai
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Mallard TT, Savage JE, Johnson EC, Huang Y, Edwards AC, Hottenga JJ, Grotzinger AD, Gustavson DE, Jennings MV, Anokhin A, Dick DM, Edenberg HJ, Kramer JR, Lai D, Meyers JL, Pandey AK, Paige Harden K, Nivard MG, de Geus EJC, Boomsma DI, Agrawal A, Davis LK, Clarke TK, Palmer AA, Sanchez-Roige S. Item-Level Genome-Wide Association Study of the Alcohol Use Disorders Identification Test in Three Population-Based Cohorts. Am J Psychiatry 2022; 179:58-70. [PMID: 33985350 PMCID: PMC9272895 DOI: 10.1176/appi.ajp.2020.20091390] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Genome-wide association studies (GWASs) of the Alcohol Use Disorders Identification Test (AUDIT), a 10-item screen for alcohol use disorder (AUD), have elucidated novel loci for alcohol consumption and misuse. However, these studies also revealed that GWASs can be influenced by numerous biases (e.g., measurement error, selection bias), which may have led to inconsistent genetic correlations between alcohol involvement and AUD, as well as paradoxically negative genetic correlations between alcohol involvement and psychiatric disorders and/or medical conditions. The authors used genomic structural equation modeling to elucidate the genetics of alcohol consumption and problematic consequences of alcohol use as measured by AUDIT. METHODS To explore these unexpected differences in genetic correlations, the authors conducted the first item-level and the largest GWAS of AUDIT items (N=160,824) and applied a multivariate framework to mitigate previous biases. RESULTS The authors identified novel patterns of similarity (and dissimilarity) among the AUDIT items and found evidence of a correlated two-factor structure at the genetic level ("consumption" and "problems," rg=0.80). Moreover, by applying empirically derived weights to each of the AUDIT items, the authors constructed an aggregate measure of alcohol consumption that was strongly associated with alcohol dependence (rg=0.67), moderately associated with several other psychiatric disorders, and no longer positively associated with health and positive socioeconomic outcomes. Lastly, by conducting polygenic analyses in three independent cohorts that differed in their ascertainment and prevalence of AUD, the authors identified novel genetic associations between alcohol consumption, alcohol misuse, and health. CONCLUSIONS This work further emphasizes the value of AUDIT for both clinical and genetic studies of AUD and the importance of using multivariate methods to study genetic associations that are more closely related to AUD.
Collapse
Affiliation(s)
- Travis T Mallard
- Department of Psychology, University of Texas at Austin, Austin, TX, 78712
| | - Jeanne E Savage
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Netherlands, 1081HV
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110
| | - Yuye Huang
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Alexis C Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, VA 23298
| | - Jouke J Hottenga
- Dept of Biological Psychology & EMGO+ Institute for Health and Care Research, Vrije Universiteit Amsterdam, Amsterdam, NL
| | | | - Daniel E Gustavson
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Andrey Anokhin
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA 23220
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - John R Kramer
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 4622
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203
| | - Ashwini K Pandey
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203
| | | | - Michel G Nivard
- Dept of Biological Psychology & EMGO+ Institute for Health and Care Research, Vrije Universiteit Amsterdam, Amsterdam, NL
| | - Eco JC de Geus
- Dept of Biological Psychology & EMGO+ Institute for Health and Care Research, Vrije Universiteit Amsterdam, Amsterdam, NL
| | - Dorret I Boomsma
- Dept of Biological Psychology & EMGO+ Institute for Health and Care Research, Vrije Universiteit Amsterdam, Amsterdam, NL
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Toni-Kim Clarke
- Division of Psychiatry, University of Edinburgh, Scotland, UK, EH8 9YL
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
31
|
Kranzler HR, Zhou H, Kember RL. Identifying and Reducing Bias in Genome-Wide Association Studies of Alcohol-Related Traits. Am J Psychiatry 2022; 179:14-16. [PMID: 34974756 DOI: 10.1176/appi.ajp.2021.21111107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Henry R Kranzler
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Kranzler, Kember); Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kranzler, Kember); Department of Psychiatry, Yale School of Medicine, New Haven, Conn., and Veterans Affairs Connecticut Healthcare System, West Haven, Conn. (Zhou)
| | - Hang Zhou
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Kranzler, Kember); Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kranzler, Kember); Department of Psychiatry, Yale School of Medicine, New Haven, Conn., and Veterans Affairs Connecticut Healthcare System, West Haven, Conn. (Zhou)
| | - Rachel L Kember
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Kranzler, Kember); Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kranzler, Kember); Department of Psychiatry, Yale School of Medicine, New Haven, Conn., and Veterans Affairs Connecticut Healthcare System, West Haven, Conn. (Zhou)
| |
Collapse
|
32
|
Gelernter J, Polimanti R. Genetics of substance use disorders in the era of big data. Nat Rev Genet 2021; 22:712-729. [PMID: 34211176 PMCID: PMC9210391 DOI: 10.1038/s41576-021-00377-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Substance use disorders (SUDs) are conditions in which the use of legal or illegal substances, such as nicotine, alcohol or opioids, results in clinical and functional impairment. SUDs and, more generally, substance use are genetically complex traits that are enormously costly on an individual and societal basis. The past few years have seen remarkable progress in our understanding of the genetics, and therefore the biology, of substance use and abuse. Various studies - including of well-defined phenotypes in deeply phenotyped samples, as well as broadly defined phenotypes in meta-analysis and biobank samples - have revealed multiple risk loci for these common traits. A key emerging insight from this work establishes a biological and genetic distinction between quantity and/or frequency measures of substance use (which may involve low levels of use without dependence), versus symptoms related to physical dependence.
Collapse
Affiliation(s)
- Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA.
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA.
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| |
Collapse
|
33
|
Mazumder AH, Barnett J, Isometsä ET, Lindberg N, Torniainen-Holm M, Lähteenvuo M, Lahdensuo K, Kerkelä M, Ahola-Olli A, Hietala J, Kampman O, Kieseppä T, Jukuri T, Häkkinen K, Cederlöf E, Haaki W, Kajanne R, Wegelius A, Männynsalo T, Niemi-Pynttäri J, Suokas K, Lönnqvist J, Tiihonen J, Paunio T, Vainio SJ, Palotie A, Niemelä S, Suvisaari J, Veijola J. Reaction Time and Visual Memory in Connection to Hazardous Drinking Polygenic Scores in Schizophrenia, Schizoaffective Disorder and Bipolar Disorder. Brain Sci 2021; 11:brainsci11111422. [PMID: 34827421 PMCID: PMC8615595 DOI: 10.3390/brainsci11111422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022] Open
Abstract
The purpose of this study was to explore the association of cognition with hazardous drinking Polygenic Scores (PGS) in 2649 schizophrenia, 558 schizoaffective disorder, and 1125 bipolar disorder patients in Finland. Hazardous drinking PGS was computed using the LDPred program. Participants performed two computerized tasks from the Cambridge Automated Neuropsychological Test Battery (CANTAB) on a tablet computer: the 5-choice serial reaction time task, or Reaction Time (RT) test, and the Paired Associative Learning (PAL) test. The association between hazardous drinking PGS and cognition was measured using four cognition variables. Log-linear regression was used in Reaction Time (RT) assessment, and logistic regression was used in PAL assessment. All analyses were conducted separately for males and females. After adjustment of age, age of onset, education, household pattern, and depressive symptoms, hazardous drinking PGS was not associated with reaction time or visual memory in male or female patients with schizophrenia, schizoaffective, and bipolar disorder.
Collapse
Affiliation(s)
- Atiqul Haq Mazumder
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Correspondence:
| | - Jennifer Barnett
- Cambridge Cognition, University of Cambridge, Cambridge CB25 9TU, UK;
| | - Erkki Tapio Isometsä
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
| | - Nina Lindberg
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
| | - Minna Torniainen-Holm
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Kaisla Lahdensuo
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
| | - Martta Kerkelä
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
| | - Ari Ahola-Olli
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Jarmo Hietala
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
- Department of Psychiatry, Turku University Hospital, 20521 Turku, Finland
| | - Olli Kampman
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland;
- Department of Psychiatry, Pirkanmaa Hospital District, 33521 Tampere, Finland
| | - Tuula Kieseppä
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
| | - Tuomas Jukuri
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Katja Häkkinen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Erik Cederlöf
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
| | - Willehard Haaki
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
| | - Risto Kajanne
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Asko Wegelius
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Teemu Männynsalo
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Social Services and Health Care Sector, City of Helsinki, 00099 Helsinki, Finland
| | - Jussi Niemi-Pynttäri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Social Services and Health Care Sector, City of Helsinki, 00099 Helsinki, Finland
| | - Kimmo Suokas
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland;
| | - Jouko Lönnqvist
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
- Department of Psychiatry, University of Helsinki, 00014 Helsinki, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Center for Psychiatry Research, Stockholm City Council, 11364 Stockholm, Sweden
| | - Tiina Paunio
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
- Department of Psychiatry, University of Helsinki, 00014 Helsinki, Finland
| | - Seppo Juhani Vainio
- Infotech Oulu, University of Oulu, 90014 Oulu, Finland;
- Northern Finland Biobank Borealis, Oulu University Hospital, 90220 Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
- Kvantum Institute, University of Oulu, 90014 Oulu, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
- Stanley Center for Psychiatric Research, The Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, MA 02142, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Solja Niemelä
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
- Department of Psychiatry, Turku University Hospital, 20521 Turku, Finland
| | - Jaana Suvisaari
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.L.); (J.S.)
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Department of Psychiatry, Oulu University Hospital, 90220 Oulu, Finland
| |
Collapse
|
34
|
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
|
35
|
Colbert SMC, Funkhouser SA, Johnson EC, Morrison CL, Hoeffer CA, Friedman NP, Ehringer MA, Evans LM. Novel characterization of the multivariate genetic architecture of internalizing psychopathology and alcohol use. Am J Med Genet B Neuropsychiatr Genet 2021; 186:353-366. [PMID: 34569141 PMCID: PMC8556277 DOI: 10.1002/ajmg.b.32874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/12/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022]
Abstract
Genetic correlations suggest that the genetic relationship of alcohol use with internalizing psychopathology depends on the measure of alcohol use. Problematic alcohol use (PAU) is positively genetically correlated with internalizing psychopathology, whereas alcohol consumption ranges from not significantly correlated to moderately negatively correlated with internalizing psychopathology. To explore these different genetic relationships of internalizing psychopathology with alcohol use, we performed a multivariate genome-wide association study of four correlated factors (internalizing psychopathology, PAU, quantity of alcohol consumption, and frequency of alcohol consumption) and then assessed genome-wide and local genetic covariance between these factors. We identified 14 significant regions of local, largely positive, genetic covariance between PAU and internalizing psychopathology and 12 regions of significant local genetic covariance (including both positive and negative genetic covariance) between consumption factors and internalizing psychopathology. Partitioned genetic covariance among functional annotations suggested that brain tissues contribute significantly to positive genetic covariance between internalizing psychopathology and PAU but not to the genetic covariance between internalizing psychopathology and quantity or frequency of alcohol consumption. We hypothesize that genome-wide genetic correlations between alcohol use and psychiatric traits may not capture the more complex shared or divergent genetic architectures at the locus or tissue specific level. This study highlights the complexity of genetic architectures of alcohol use and internalizing psychopathology, and the differing shared genetics of internalizing disorders with PAU compared to consumption.
Collapse
Affiliation(s)
- Sarah M. C. Colbert
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder
| | | | - Emma C. Johnson
- Department of Psychiatry, Washington University School of Medicine
| | - Claire L. Morrison
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Charles A. Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Integrative Physiology, University of Colorado Boulder
| | - Naomi P. Friedman
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Marissa A. Ehringer
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Integrative Physiology, University of Colorado Boulder
| | - Luke M. Evans
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder
| |
Collapse
|
36
|
Mazumder AH, Barnett J, Isometsä ET, Lindberg N, Torniainen-Holm M, Lähteenvuo M, Lahdensuo K, Kerkelä M, Ahola-Olli A, Hietala J, Kampman O, Kieseppä T, Jukuri T, Häkkinen K, Cederlöf E, Haaki W, Kajanne R, Wegelius A, Männynsalo T, Niemi-Pynttäri J, Suokas K, Lönnqvist J, Tiihonen J, Paunio T, Vainio SJ, Palotie A, Niemelä S, Suvisaari J, Veijola J. Reaction Time and Visual Memory in Connection to Alcohol Use in Persons with Bipolar Disorder. Brain Sci 2021; 11:brainsci11091154. [PMID: 34573174 PMCID: PMC8467646 DOI: 10.3390/brainsci11091154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study was to explore the association of cognition with hazardous drinking and alcohol-related disorder in persons with bipolar disorder (BD). The study population included 1268 persons from Finland with bipolar disorder. Alcohol use was assessed through hazardous drinking and alcohol-related disorder including alcohol use disorder (AUD). Hazardous drinking was screened with the Alcohol Use Disorders Identification Test for Consumption (AUDIT-C) screening tool. Alcohol-related disorder diagnoses were obtained from the national registrar data. Participants performed two computerized tasks from the Cambridge Automated Neuropsychological Test Battery (CANTAB) on A tablet computer: the 5-choice serial reaction time task, or reaction time (RT) test and the Paired Associative Learning (PAL) test. Depressive symptoms were assessed with the Mental Health Inventory with five items (MHI-5). However, no assessment of current manic symptoms was available. Association between RT-test and alcohol use was analyzed with log-linear regression, and eβ with 95% confidence intervals (CI) are reported. PAL first trial memory score was analyzed with linear regression, and β with 95% CI are reported. PAL total errors adjusted was analyzed with logistic regression and odds ratios (OR) with 95% CI are reported. After adjustment of age, education, housing status and depression, hazardous drinking was associated with lower median and less variable RT in females while AUD was associated with a poorer PAL test performance in terms of the total errors adjusted scores in females. Our findings of positive associations between alcohol use and cognition in persons with bipolar disorder are difficult to explain because of the methodological flaw of not being able to separately assess only participants in euthymic phase.
Collapse
Affiliation(s)
- Atiqul Haq Mazumder
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Correspondence:
| | - Jennifer Barnett
- Cambridge Cognition, University of Cambridge, Cambridge CB25 9TU, UK;
| | - Erkki Tapio Isometsä
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Department of Psychiatry, University of Helsinki, 00014 Helsinki, Finland;
| | - Nina Lindberg
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
| | - Minna Torniainen-Holm
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Kaisla Lahdensuo
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
| | - Martta Kerkelä
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
| | - Ari Ahola-Olli
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Jarmo Hietala
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
- Department of Psychiatry, Turku University Hospital, 20521 Turku, Finland
| | - Olli Kampman
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland;
- Department of Psychiatry, Pirkanmaa Hospital District, 33521 Tampere, Finland
| | - Tuula Kieseppä
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
| | - Tuomas Jukuri
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Katja Häkkinen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Erik Cederlöf
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
| | - Willehard Haaki
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
| | - Risto Kajanne
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Asko Wegelius
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
| | - Teemu Männynsalo
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Social Services and Health Care Sector, City of Helsinki, 00099 Helsinki, Finland
| | - Jussi Niemi-Pynttäri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Social Services and Health Care Sector, City of Helsinki, 00099 Helsinki, Finland
| | - Kimmo Suokas
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland;
| | - Jouko Lönnqvist
- Department of Psychiatry, University of Helsinki, 00014 Helsinki, Finland;
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
| | - Jari Tiihonen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, 70240 Kuopio, Finland; (M.L.); (K.H.); (J.T.)
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Center for Psychiatry Research, Stockholm City Council, 11364 Stockholm, Sweden
| | - Tiina Paunio
- Department of Psychiatry, University Hospital and University of Helsinki, 00029 Helsinki, Finland; (E.T.I.); (N.L.); (T.K.); (A.W.); (T.P.)
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
- Social Services and Health Care Sector, City of Helsinki, 00099 Helsinki, Finland
| | - Seppo Juhani Vainio
- Infotech Oulu, University of Oulu, 90014 Oulu, Finland;
- Northern Finland Biobank Borealis, Oulu University Hospital, 90220 Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
- Kvantum Institute, University of Oulu, 90014 Oulu, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland; (K.L.); (A.A.-O.); (W.H.); (R.K.); (T.M.); (J.N.-P.); (K.S.); (A.P.)
- Mehiläinen, Pohjoinen Hesperiankatu 17 C, 00260 Helsinki, Finland
- Stanley Center for Psychiatric Research, The Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, MA 02142, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Solja Niemelä
- Department of Psychiatry, University of Turku, 20014 Turku, Finland; (J.H.); (S.N.)
- Department of Psychiatry, Turku University Hospital, 20521 Turku, Finland
| | - Jaana Suvisaari
- Mental Health Unit, Finnish Institute for Health and Welfare (THL), 00271 Helsinki, Finland; (M.T.-H.); (E.C.); (J.S.)
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, 90014 Oulu, Finland; (M.K.); (T.J.); (J.V.)
- Department of Psychiatry, Oulu University Hospital, 90220 Oulu, Finland
| |
Collapse
|
37
|
Dissecting polygenic signals from genome-wide association studies on human behaviour. Nat Hum Behav 2021; 5:686-694. [PMID: 33986517 DOI: 10.1038/s41562-021-01110-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/31/2021] [Indexed: 02/03/2023]
Abstract
Genome-wide association studies on human behavioural traits are producing large amounts of polygenic signals with significant predictive power and potentially useful biological clues. Behavioural traits are more distal and are less directly under biological control compared with physical characteristics, which makes the associated genetic effects harder to interpret. The results of genome-wide association studies for human behaviour are likely made up of a composite of signals from different sources. While sample sizes continue to increase, we outline additional steps that need to be taken to better delineate the origin of the increasingly stronger polygenic signals. In addition to genetic effects on the traits themselves, the major sources of polygenic signals are those that are associated with correlated traits, environmental effects and ascertainment bias. Advances in statistical approaches that disentangle polygenic effects from different traits as well as extending data collection to families and social circles with better geographical coverage will probably contribute to filling the gap of knowledge between genetic effects and behavioural outcomes.
Collapse
|
38
|
Using Genetic Marginal Effects to Study Gene-Environment Interactions with GWAS Data. Behav Genet 2021; 51:358-373. [PMID: 33899139 DOI: 10.1007/s10519-021-10058-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 04/09/2021] [Indexed: 12/30/2022]
Abstract
Gene-environment interactions (GxE) play a central role in the theoretical relationship between genetic factors and complex traits. While genome wide GxE studies of human behaviors remain underutilized, in part due to methodological limitations, existing GxE research in model organisms emphasizes the importance of interpreting genetic associations within environmental contexts. In this paper, we present a framework for conducting an analysis of GxE using raw data from genome wide association studies (GWAS) and applying the techniques to analyze gene-by-age interactions for alcohol use frequency. To illustrate the effectiveness of this procedure, we calculate genetic marginal effects from a GxE GWAS analysis for an ordinal measure of alcohol use frequency from the UK Biobank dataset, treating the respondent's age as the continuous moderating environment. The genetic marginal effects clarify the interpretation of the GxE associations and provide a direct and clear understanding of how the genetic associations vary across age (the environment). To highlight the advantages of our proposed methods for presenting GxE GWAS results, we compare the interpretation of marginal genetic effects with an interpretation that focuses narrowly on the significance of the interaction coefficients. The results imply that the genetic associations with alcohol use frequency vary considerably across ages, a conclusion that may not be obvious from the raw regression or interaction coefficients. GxE GWAS is less powerful than the standard "main effect" GWAS approach, and therefore require larger samples to detect significant moderated associations. Fortunately, the necessary sample sizes for a successful application of GxE GWAS can rely on the existing and on-going development of consortia and large-scale population-based studies.
Collapse
|